Skip to main content

Full text of "USPTO Patents Application 09869475"

See other formats


WORLD INTEUXCTUAL PROPERTY ORGANIZATION 

International Bureac 




PCX 

INTERNATIONAL APPUCATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 



(51) International Patent ClassiCcatlon ^ : 

A61K 38A9, 38/18, 4m0, A61P 25/02 



A2 



(11) InternaUonal PubUcatlon Number: WO 00/62798 

(43) International PubUcation Date: 26 October 2000 (26.10.00) 



(21) Internaaonal Application Number: PCT/US00/(^765 

(22) International Filing Date: 1 1 April 2000 (1 1 .04.00) 



(30) Priority Data: 
60/129,768 



15 April 1999 (15.04.99) 



US 



(71) Applicant: ST. ELIZABETH'S MEDICAL CENTER. INC. 

[US/US]; 736 Cambridge Street, Boston, MA 02135 (US). 

(72) Inventor: ISNER, Jeffrey. M.; 34 Brenton Road, Weston, MA 

02193 (US). 

(74) Agent: PLUMER, Elizabeth, R.; Wolf. Greenfield & Sacks. 
P.C., 600 Atlantic Avenue. Boston. MA 02210 (US). 



(81) Designated States: AU. CA. JP. European patent (AT, BE, 
CH, CY, DE, DK. ES. FI. PR. GB. GR, IE. IT. LU, MC, 
NL, PT. SE). 



Published 

Without international search report and to be republished 
upon receipt of that report. 



(54) TiUe: ANGIOGENIC GROWTH FACTORS FOR TREATMENT OF PERIPHERAL NEUROPATHY 
(57) Abstract 

A me^CKt for treating peripheral neuropathy, particularly ischemic {^ripheral neuropathy, is provided. The method involves 
administering to subjects in need of such treatment an effective amount of an angiogenic growth factor to alleviate a symptom of the 
neuropathy. 



FOR THE PURPOSES OF INFORMATION ONLY 



Codes used to identify States party to the PCX on the front pages of pamphlets publishing international applications under the PCX. 



AL 


Albania 


£S 


Spain 


LS 


Lesotho 


SI 


Slovenia 


AM 


Annenia 


n 


Finland 


LT 


Lithuania 


SK 


Slovakia 


AT 


Austria 


FR 


France 


LU 


Luxembourg 


SN 


Senegal 


AU 


Australia 


GA 


Gabon 


LV 


Latvia 


SZ 


Swaziland 


AZ 


Azerbaijan 


GB 


United Kingdom 


MC 


Monaco 


TD 


Chad 


BA 


Bosnia and Herzegovina 


GE 


Georgia 


MD 


Republic of Moldova 


TG 


Togo 


BB 


Barbados 


GH 


Ghana 


MG 


Mad^ascar 


TJ 


Tajikistan 


BE 


Belgium 


GN 


Guinea 


MK 


The fonncT Yugoslav 


TM 


'nnkmenistan 


BF 


Burkina Faso 


GR 


Greece 




Republic of Macedonia 


TR 


Tmkcy 


BG 


Bulgaria 


HU 


Hungary 


ML 


Mali 


TT 


Trinidad and Tobago 


BJ 


Benin 


IE 


Ireland 


MN 


Mongolia 


UA 


Ukraine 


BR 


Brazil 


IL 


Israel 


MR 


Mauritania 


UG 


Uganda 


BY 


Belaius 


IS 


Iceland 


MW 


Malawi 


US 


United Stales of America 


CA 


Canada 


IT 


Italy 


MX 


Mexico 


UZ 


Uzbekistan 


CF 


Central African Republic 


J? 


Japan 


NE 


Niger 


VN 


Viet Nam 


CG 


Congo 


KE 


Kenya 


NL 


Netherlands 


YU 


Yugoslavia 


CH 


Switzerbtnd 


KG 


Kyrgyzstan 


NO 


Norway 


ZW 


Zimbabwe 


a 


Cdic d'lvone 


KP 


Democratic People*s 


NZ 


New Zealand 






CM 


Cameroon 




Republic of Korea 


PL 


Poland 






CN 


China 


KR 


Republic of Korea 


PT 


Portugal 






CU 


Cuba 


KZ 


Kazakstan 


RO 


Romania 






CZ 


Ct^ch Republic 


VC 


Saim Luda 


RU 


Rosstan Fbderaiion 






DE 


Germany 


U 


Liechtenstein 


SD 


Sudan 






DK 


Denmark 


LK 


Sri Lanka 


SB 


Sweden 






EE 


Estonia 


LR 


Liberia 


SG 


Smgapore 







f 

wo 00/62798 PCT/USOO/09765 

-1- 

ANGIOGENIC GROWTH FACTORS 
FOR TREATMENT OF PERIPHERAL NEUROPATHY 
Field of the Invention 

This invention relates to methods and compositions for the treatment of peripheral 
5 neuropathy, such as ischemic peripheral neuropathy. The methods involve administering 
an angiogenic growth factor to alleviate a symptom of a peripheral neuropathy and, 
optionally, enhance nerve regeneration in a mammal. 

Background of the Invention 
Peripheral neuropathy generally refers to a disorder that affects the peripheral 

10 nerves, most often manifested as one or a combination of motor, sensory, sensorimotor, 
or autonomic neural dysfimction. Peripheral neuropathies can be genetically acquired, 
can be induced by a toxic agent, or can result from peripheral ischemia or from a 
systemic disease. Genetically-acquired peripheral neuropathies include Refsum's 
disease, Abetalipoproteinemia, Tangier disease, Krabbe's disease. Metachromatic 

15 leukodystrophy, Fabry's disease, Dejerine-Sottas syndrome, Charcot-Marie Tooth 
Disease (also known as Peroneal Muscular Atrophy, or Hereditary Motor Sensory 
Neuropathy) and others. Exemplary toxic agents which cause neurotoxicities include 
therapeutic drugs such as antineoplastic agents, contaminants in foods, and environmental 
and mdustrial pollutants. Ischemic peripheral neuropathies include, but are not limited to, 

20 diabetic polyneuropathy. Peripheral neuropathies associated with a systemic condition 
include uremia and alcoholic polyneuropathy among other disorders. 

Ischemic peripheral neuropathy, particularly when it develops in the absence of 
diabetes, has received limited study, despite the fact that it may be a prominent feature of 
patients with peripheral vascular disease. Among diabetics, peripheral neuropathy is 

25 common and ultimately accounts for significant morbidity. Typically, symptoms are 
dominated by sensory defects. (Tomlinson DR, et al. Diabetes 1997;46:S43-S49). The 
ultimate consequence of such sensory deficits involving the lower extremities may be 
foot ulceration initiated by traumatic injury that is inapparent to the patient. Indeed, it has 
been reported that 20% of all hospital admissions among diabetic patients in the United 

30 States are for foot problems. (Reiber GE, et al., in Harris MI, et al, (eds): Diabetes in 

America. Washington, National Institute of Diabetes and Digestive and Kidney Diseases, 



wo 00/62798 



-2- 



PCT/USOO/09765 



1995, pp 409-427). That such ulcerations may lead to lower extremity amputation 
(Parkhouse N, et al,,NEnglJMed 1988;31 8:1306-1309) is borne out by the fact that the 
rate of lower limb amputation is fifteen times higher in diabetic versus non-diabetic 
patients (Veves A, et al., Diabetes 1998;47:457-463). Even with intensive insulin 
5 therapy, as reported in the Diabetes Control and Complications Trial (DCCT), the 

incidence of new clinically detected neuropathy per patient per year was 3.1% in the non- 
retinopathy group and 7.0% in the group with baseline retinopathy; with conventional 
therapy, the incidence of neuropathy increased to 9.8% and 16.1% with and without 
retinopathy (The Diabetes Control and Complications Trial Research Group, N Engl J 

10 Med 1993;329:977-988). When loss of sensation is compounded by loss of control over 
blood flow due to autonomic neuropathy and lower extremity vascular obstruction, the 
threat of limb loss is exacerbated. In the case of peripheral artery disease, hospital 
mortality, length of hospitalization, and complications resulting from surgery are all 
increased in the presence of diabetes (Currie CJ, et al., Diabetes Care 1998;21:42-48). 

15 In view of the foregoing, a need still exists to better understand the molecular 

processes underlying peripheral neuropathy, and to develop improved drug therapies to 
replace or supplement the existing methods for treating peripheral neuropathies, 
particularly ischemic peripheral neuropathy. Preferably, such drug therapies would be 
designed to reduce or prevent nerve damage at its earliest stages and to enhance 

20 peripheral nerve repair following diagnosis and treatment. 

Summary of the Invention 
The invention is based, in part, on the observation that a nimiber of patients 
receiving angiogenic growth factor treatment for vascular insufficiency appeared to 
exhibit an improvement in sensory neuropathy. To test Applicant's hypothesis that the 

25 angiogenic grov^ factor either directly or indirectly improved the peripheral neuropathy. 
Applicant developed an animal model of hindlimb ischemia which exhibits severe 
peripheral neuropathy and which, in contrast to existing in vitro and in vivo methods, is 
predictive of an in vivo therapeutic effect of an agent for treating a peripheral neuropathy, 
administered (intramuscularly) to this animal model a vector containing a nucleic acid 

30 encoding VEGF ("VEGF vector"), and discovered that this angiogenic growth factor 

attenuated the development of ischemic peripheral neuropathy and enhanced the recovery 
of established ischemic peripheral neuropathy. Thus, Applicant describes herein newly 



wo 00/62798 



-3- 



PCTA;S00/09765 



discovered functions for VEGF and other angiogenic growth factors, namely, the ability 
to prevent or reduce ischemia induced nerve damage at its earliest stages and the ability 
to enhance peripheral nerve repair following the onset of peripheral neuropathy. 
Accordingly, the instant invention is directed to compositions and methods that are based 
5 upon the discovery of these newly-discovered functions for angiogenic growth factors. 
Exemplary conditions that are characterized by peripheral neuropathy include: (a) an 
ischemic peripheral neuropathy; (b) a neuropathy associated with a systemic condition; 
(c) a toxin-induced peripheral neuropathy; and (d) a genetically acquired peripheral 
neuropathy. 

10 According to one aspect of the invention, a method for treating a condition 

characterized by peripheral neuropathy in a subject is provided. In certeiin preferred 
embodiments, the subject is otherwise free of symptoms calling for treatment with an 
angiogenic growth factor. The method involves administering at least one angiogenic 
growth factor (an "angiogenic growth factor nucleic acid" or an "angiogenic growth 

15 factor polypeptide") to a subject in need of such treatment in an amount effective to 

alleviate a symptom of peripheral neuropathy in the subject. The symptom of peripheral 
neuropathy can be one or more of the symptoms which are used by the skilled medial 
professional to diagnose a peripheral neuropathy. 

Exemplary angiogenic growth factors (including all genes and isoforms of each 

20 gene product) for use in accordance with the methods of the invention include: vascular 
endothelial cell growth factor (VEGF), acidic fibroblast growth factor (aFGF), basic 
fibroblast growth factor (bFGF), epidermal growth factor, transforming growth factors a 
and P, platelet-derived endothelial growth factor, platelet-derived growth factor, tumor 
necrosis factor a, hepatocyte growth factor (scatter factor), erythropoietin, colony 

25 stimulating factor (CSF), macrophage-CSF(M-CSF), granulocyte/macrophage CSF (GM- 
CSF), angiopoietin 1 and 2, and nitric oxide synthase (NOS). The nucleic acid and amino 
acid sequences for these and other angiogenic growth factors are available in public 
databases such as GenBank and in the literature. The preferred angiogenic growth factor 
is VEGF, more preferably, a VEGF nucleic acid. In certain particularly preferred 

30 embodiments, the VEGF nucleic acid is administered to the subject in conjunction ynth a 
second angiogenic growth factor nucleic acid which, preferably, is bFGF. The 
compositions and methods of the invention are useful for replacing existing drug 



wo 00/62798 



-4- 



PCTAJSQO/09765 



therapies, as well as for improving the effectiveness of existing therapies for treating 
conditions that are characterized by peripheral neuropathy. In general, such conditions 
are diagnosed by detecting one or a combination of motor, sensory, sensorimotor, or 
autonomic neural dysfunctions in the subject. 

5 An "angiogenic growth factor" embraces an "angiogenic growth factor nucleic 

acid" and an "angiogenic growth factor polypeptide". As used herein, an "angiogenic 
growth factor polypeptide" refers to any protein, polypeptide, mutein or portion thereof 
that is capable of inducing endothelial cell growth. VEGF is a particularly preferred 
angiogenic growth factor (e.g., VEGF 1 (also referred to as VEGF A); VEGF 2 (also 

10 referred to as VEGF C); VEGF B; and VEGF D). An angiogenic growth factor nucleic 
acid refers to a nucleic acid which encodes an angiogenic growth factor polypeptide. The 
invention embraces the administration of angiogenic growth factor nucleic acids and 
polypeptides for the treatment of peripheral neuropathies. The invention also embraces 
agents that upregulate expression of an angiogenic growth factor polypeptide in vivo. 

15 The complete coding sequence for representative preferred human angiogenic 

growth factors cDNA and predicted amino acid sequence are available in public 
databases such as GenBank and in literature. In particular, certain of the VEGF genes, 
isoforms, fragments, and analogs thereof that are useful for practicing the claimed 
invention are described in GenBank Accession Nos. NM 003376 ("Homo sapiens 

20 vascular endothelial growth factor (VEGF) mRNA"); NM 003377 ("Homo sapiens 
vascular endothelial growth factor B (VEGFB) mRNA"); NM 005429 ("Homo sapiens 
vascular endothelial growth factor C (VEGFC) mRNA"); NM 004469 ("Homo sapiens c- 
fos induced growth factor (vascular endothelial growth factor D) (FIGF) mRNA); AF 
024710 ("Homo sapiens vascular growth factor (VEGF (165)) mRNA, 3'UTR, mRNA 

25 sequence"); and U.S. Patent Nos. 6,013,780 ("VEGF145 expression vectors"); 5,935,820 
("Polynucleotides encoding vasctdar endodielial growth factor 2"); 5,607,918 ("Vascular 
endothelial growth factor-B and DNA coding therefor"); and 5,219,739 ("DNA 
sequences encoding bVEGF 120 and hVEGF 121 and methods for the production of 
bovine and human vascular endothelial cell growth factors, bVEGFi2o and hVEGF^i"), 

30 including references cited therein, the entire contents of the foregoing accession numbers, 
patent docimients, and references are incorporated in their entirety by reference. 



wo 00/62798 



PCT/USOO/09765 



-5- 

The preferred angiogenic growth factor nucleic acids of the invention encode the 
above-identified angiogenic growth factor polypeptides, as well as their homologs and 
alleles and functionally equivalent fi-agments or variants of the foregoing. For example, 
human VEGF 1 (VEGF A) exists in four principal isoforms, phVEGFi2i; phVEGF]45; 
5 phVEGFi65; and phVEGFigg. Preferably, the angiogenic growth factor nucleic acid has 
the nucleotide sequence encoding an intact human angiogenic growth factor polypeptide, 
i.e., the complete coding sequence of the gene encoding a human angiogenic grovs^ 
factor; however the invention also embraces the use of nucleic acids encoding fi-agments 
of an intact angiogenic growth factor. 

10 In general, the angiogenic growth factor nucleic acid is operatively coupled to a 

promoter that can express the angiogenic growth factor in a targeted cell (e.g., an 
endothelial cell, a nerve cell, a muscle cell). Optionally, the nucleic acid is contained in 
an appropriate expression vector (e.g., plasmid, adenoviral vector, modified adenoviral 
vector, retroviral vector, liposome) to more efficiently genetically modify the targeted 

15 cell and achieve expression of the angiogenic growth factor. 

According to another aspect of the invention, an alternative method for treating a 
condition characterized by peripheral nexiropathy in a subject is provided. The method 
involves: administering at least one angiogenic growth factor to a subject in need of such 
treatment in an amount and for a period of time effective to alleviate a symptom of 

20 peripheral neuropathy in the subject. The symptom of peripheral neuropathy can be one 
or more of the symptoms which are used by the skilled medical professional to diagnose a 
peripheral neuropathy. The period of time that is effective for alleviating a symptom of 
peripheral neuropathy is significantly greater than the period of time during which an 
angiogenic growth factor typically is administered to a subject for the purpose of 

25 revascularization in an ischemic tissue. In general, angiogenic growth factors are 
administered to a patient for a period of up to, and including, about twelve weeks to 
enhance blood vessel development in ischemic tissue. In contrast, in the preferred 
embodiments of this aspect of the invention, administration of the angiogenic growth 
factor is for greater than twelve weeks; more preferably, greater than eighteen weeks; and 

30 most preferably, greater than about twenty-four weeks. In some instances, treatment for 
the pmposes of this aspect of the invention is continued for at least six months to several 



wo 00/62798 



PCT/USOO/09765 



-6- 

years, and more preferably, from six months to one, two, three years or for the patient's 
lifetime in the case of chronic peripheral neuropathy. 

According to yet another aspect of the invention, a further method for treating a 
condition characterized by peripheral neuropathy in a subject is provided. The method 
5 involves administering at least one angiogenic growth factor to a subject in need of such 
treatment in an amount effective to alleviate a symptom of peripheral neuropathy in the 
subject, wherein administering is by intramuscular injection into a tissue at an injection 
site that is proximate to a nerve which is suspected of a neuropathy and that is distal to an 
injection site that would be selected for the puqDOse of revascularization (e.g., 

10 neovascularization). In general, intramuscular injection into a tissue for the purpose of 
revascularization in ischemic tissue is accomplished by localized delivery of the 
angiogenic growth factor to the site of a vascular blockage. In contrast, according to the 
embodiments in which a subject also presents with a vascular insufficiency, intramuscular 
injection of angiogenic growth factors, preferably, is into the tissue at a location which 

15 excludes these locations. The preferred locations into which the angiogenic factors are 
intramuscularly injected for the purpose of treating peripheral neuropathy include sites 
which are proximate to a nerve which is suspected of a neuropathy. An amount of the 
angiogenic growth factor is administered to alleviate a symptom of a peripheral 
neuropathy. The symptom of peripheral neuropathy can be one or more symptoms which 

20 are used by the skilled medical professional to diagnose a peripheral neuropathy. 

According to yet another aspect of the invention, a method for treating a subject 
who has sustained a peripheral nerve injury is provided. The method involves 
administering at least one angiogenic growth factor to a subject in need of such treatment 
in an amount effective to enhance peripheral nerve regeneration. Preferably, the subject 

25 is otherwise free of symptoms calling for treatment vwth an angiogenic growth factor. 
The angiogenic growth factor nucleic acids and polypeptides and exemplary conditions 
which are characterized by peripheral neuropathy are as described above. 

It is to be understood that an angiogenic growth factor polypeptide can be used in 
place of an angiogenic growth factor nucleic acid in treating any of the foregoing 

30 conditions. Thus, according to still another aspect of the invention, pharmaceutical 
preparations are provided that contain an angiogenic growth factor nucleic acid or an 
angiogenic growth factor polypeptide. The pharmaceutical preparations contain the 



wo 00/62798 



PCT/USOO/09765 



-7- 

above-described angiogenic growth factors, together with a pharmaceutically-acceptable 
carrier. Preferably, the angiogenic growth factors are present in the compositions in an 
amount effective for treating a peripheral neuropathy. The angiogenic growth factors are 
particularly useful for the treatment of ischemic peripheral neuropathy. Preferably, this 
5 amoimt is sufficient to enhance nerve regeneration in vivo. 

According to still another aspect of the invention, the above angiogenic growth 
factors (angiogenic growth factor nucleic acids and angiogenic growth factor 
polypeptides), alone or in combination, are used in the preparation of medicaments for 
the treatment of a peripheral neuropathy. The method involves placing the angiogenic 

10 growth factor(s) in a pharmaceutically-acceptable carrier. The preferred angiogenic 
growth factors are as described above. 

It is noteworthy that in certain embodiments, the preferred subjects treated 
according to the methods set forth above are otherwise free of symptoms calling for 
angiogenic growth factor treatment, either by administration of the angiogenic growth 

15 factor polypeptide or by an angiogenic growth factor nucleic acid. Thus, in certain select 
embodiments, the subjects are not otherwise being treated using a gene therapy protocol 
or, if being treated using gene therapy, the protocol for the methods of the invention 
differ in the dosage or duration of treatment (greater than about twelve weeks) and/or the 
site of intramuscular injection (in the proximity of a nerve suspected of a neuropathy). 

20 The invention also contemplates the use of angiogenic growth factors in 

experimental model systems to determine the role that angiogenic growth factors play in 
the repair of peripheral nerves or in mediating an adverse health consequence occurring 
as a result of peripheral neuropathy. An ischemic animal model of peripheral neuropathy 
is described in the Examples and can be used to select agents for treatment of this 

25 condition. The agent (e.g., an angiogenic growth factor as described above) is 
administered to the animal, locally or systemically, and the animal's response is 
monitored and compared to control animals that do not receive the angiogenic growth 
factors. In this manner, additional agents which are useful for treating peripheral 
neuropathies can be identified. 

30 These and other aspects of the invention will be described in greater detail below. 

Throughout this disclosure, all technical and scientific terms have the same meaning as 



wo 00/62798 PCT/USOO/09765 

-8- 

commonly understood by one of ordinary skill in the art to which this invention pertains 
unless defined otherwise. 

Detailed Description of the Drawings 

Figure lA shows the motor nerve conduction velocity for non-diabetic and 
5 diabetic animals which received saline or VEGF; and 

Figure IB shov^ sensory nerve conduction velocity for non-diabetic and diabetic 
animals which received saline and VEGF. 

This application, particularly the Examples, may refer to Figures; however, none 
of the figures are essential for enablement of the invention. 
10 Detailed Description of the Invention 

The invention is based, in part, on the observation that a number of patients 
receiving an angiogenic growth factor to treat a vascular insufficiency appeared to exhibit 
an improvement in sensory neuropathy. Accordingly, Applicant hypothesized that the 
angiogenic growth factor either directly or indirectly improved the peripheral neuropathy. 
15 To test this hypothesis. Applicant developed an animal model of hindlimb ischemia 
which exhibits severe peripheral neuropathy and which resembles and is predictive of a 
human peripheral neuropathy. Applicant then administered (intramuscularly) to the 
animal model, a vector containing a nucleic acid encoding VEGF ("VEGF vector"), and 
discovered that this angiogenic growth factor attenuated the development of ischemic 
20 peripheral neuropathy and enhanced the recovery of established ischemic peripheral 
neuropathy. Based on these discoveries. Applicant describes herein new functions for 
VEGF and other angiogenic growth factors, namely, the ability to prevent or reduce 
ischemia induced nerve damage at its earliest stages as well as the ability to enhance 
peripheral nerve repair. Accordingly, the instant invention is directed to compositions 
25 and methods that are based upon the discovery of these newly-discovered fiinctions for 
angiogenic growth factors. 

As used herein, "peripheral neuropathy" refers to a disorder affecting a segment 
of the peripheral nervous system. The invention involves using an angiogenic growth 
factor to reduce a neuropathology including, but not limited to, a distal sensorimotor 
30 neuropathy, or an autonomic neuropathy such as reduced motility of the gastrointestinal 
tract or atony of the urinary bladder. Preferred neuropathies that can be treated with the 
angiogenic growth factors of the invention also include neuropathies associated with 



wo 00/62798 



PCT/USOO/09765 



-9- 

ischemic disease, neuropathies associated with a systemic disease, e.g., post-polio 
syndrome; genetically acquired neuropathies, e.g., Charcot-Marie-Tooth disease; and 
neuropathies caused by a toxic agent, e.g., a chemotherapeutic agent, such as vincristine. 
Each of these categories of conditions is discussed in more detail below. 
5 According to one aspect of the invention, a method for treating a condition 

characterized by peripheral neuropathy in a subject is provided. The method involves 
administering at least one angiogenic growth factor (an "angiogenic growth factor nucleic 
acid" or an "angiogenic growth factor polypeptide") to a subject in need of such treatment 
in an amount effective to alleviate a symptom of peripheral neuropathy in the subject. 

10 The symptom of peripheral neuropathy can be one or more of the symptoms which are 
used by the skilled medical professional to diagnose a peripheral neuropathy. Exemplary 
angiogenic growth factors for use in accordance with the methods of the invention are 
described below. The nucleic acid and amino acid sequences for these and other 
angiogenic growth factors are available in public databases such as GenBank and in the 

15 literature. Preferably, the subject is otherwise free of symptoms calling for treatment 
with an angiogenic growth factor. 

According to another aspect of the invention, an alternative method for treating a 
condition characterized by peripheral neuropathy in a subject is provided. The method 
involves: administering at least one angiogenic growth factor to a subject in need of such 

20 treatment in an amount and for a period of time effective to alleviate a symptom of 

peripheral neuropathy in the subject. The symptom of peripheral neuropathy can be one 
or more of the symptoms which are used by the skilled medical professional to diagnose a 
peripheral neuropathy. The period of time that is effective for alleviating a symptom of 
peripheral neuropathy is significantly greater than the period of time during which an 

25 angiogenic growth factor is administered to a subject for the purpose of revascularization 
of an ischemic tissue. Typically, angiogenic growth factors are administered to a patient 
for a period of up to, and including, about twelve weeks for enhancing blood vessel 
development in ischemic vascular tissue. In contrast, in the preferred embodiments of 
this aspect of the invention, administration of the angiogenic growth factor is for greater 

30 than about twelve weeks; more preferably, greater than eighteen weeks; and most 

preferably, greater than about twenty-four weeks. In some instances, treatment for the 
purposes of this aspect of the invention is continued for at least six months to several 



wo 00/62798 PCT/USOO/09765 

-10- 

years, and more preferably, from six months to one, two, three years, or for the patient's 
lifetime in the case of chronic peripheral neuropathy. Repeated injections using 
controlled release microparticles containing the angiogenic growth factors can be used for 
this purpose. 

5 According to yet another aspect of the invention, a further method for treating a 

condition characterized by peripheral neuropathy in a subject is provided. The method 
involves administering at least one angiogenic grov^ factor to a subject in need of such 
treatment in an amount effective to alleviate a symptom of peripheral neuropathy in the 
subject, wherein administering is by intramuscular injection into a tissue at an injection 

10 site that is proximate to a nerve which is suspected of a neuropathy and that is distal to an 
injection site that would be selected for the purpose of revascularization (e.g., 
neovascularization) in an ischemic tissue. In general, intramuscular injection into a tissue 
for the purpose of revascularization in ischemic tissue is accomplished by localized 
delivery of the angiogenic growth factor to the site of a vascular blockage. In contrast, 

15 according to the embodiments in which a subject also presents v^th a vascular 

insufficiency, intramuscular injection of angiogenic grov^ factors, preferably, is into a 
tissue at a location which excludes these locations. The locations into which the 
angiogenic factors are intramuscularly injected for the purpose of treating peripheral 
neuropathy include muscle which is proximate to a nerve which is suspected of a 

20 neuropathy. 

According to yet another aspect of the invention, a method for treating a subject 
who has sustained a peripheral nerve injury is provided. The method involves 
administering at least one angiogenic growth factor to a subject in need of such treatment 
in an amount effective to enhance peripheral nerve regeneration. Preferably, the subject 

25 is otherwise free of symptoms calling for treatment v^th an angiogenic growth factor. 

As used herein, an "angiogenic growth factor" embraces an "angiogenic growth 
factor nucleic acid" and an "angiogenic growth factor polypeptide". An "angiogenic 
grov^ factor polypeptide" refers to any protein, polypeptide, mutein or portion that is 
capable of inducing endothelial cell grov^. VEGF is a particularly preferred angiogenic 

30 growth factor. An angiogenic growth factor nucleic acid refers to a nucleic acid which 
encodes an angiogenic growth factor. The invention embraces the administration of 
nucleic acids or polypeptides for the treatment of peripheral neuropathies. 



wo 00/62798 



PCT/USOO/09765 



-11- 

Homologs and alleles of the nucleic acid and amino acid sequences reported for 
the angiogenic growth factors, such as those identified herein, also are embraced within 
the definition of an "angiogenic growth factor". In addition, the angiogenic growth 
factor nucleic acids of the invention include nucleic acids which code for the angiogenic 
5 growth factor polypeptides having the sequences reported in the public databases and/or 
literature, but which differ fi-om the naturally occurring nucleic acid sequences in codon 
sequence due to the degeneracy of the genetic code. The invention also embraces 
isolated functionally equivalent fragments, variants, and analogs of the foregoing nucleic 
acids; proteins and peptides coded for by any of the foregoing nucleic acids; and 

10 complements of the foregoing nucleic acids. Particularly preferred fragments of the 
VEGF nucleic acid and VEGF polypeptides are identified below. 

The angiogenic growth factor nucleic acid may be any nucleic acid (DNA or 
RNA) including genomic DNA, cDNA and mRNA, encoding an angiogenic growth 
factor which can be used to express a growth factor, e.g., a protein, polypeptide, mutein 

15 or portion thereof, that is capable of inducing either directly or indirectly, the formation 
of new blood vessels (Folkman, et al,, Science, 235:442-447 (1 987)). These include, for 
example, vascular endothelial growth factor (VEGF), acidic fibroblast growth factor 
(aFGF) (Bjomsson, et al., Proc. Natl. Acad. Sci. USA, 88:8651-8655, (1991)), basic 
fibroblast growth factor (bFGF) (Schwarz, et al., J. Vase Surg., 5:280-288, (1987)), 

20 epidermal growth factor (EGF), transforming grov^h factors a and P (TGF-a and TGF-P), 
platelet-derived endothelial cell growth factor (PD-ECGF), platelet-derived growth factor 
(PDGF) itself, tumor necrosis factor a (TNF-a), hepatocyte growth factor (HGF), 
erythropoietin, colony stimulating factor (CSF), macrophage-CSF(M-CSF), 
granulocyte/macrophage CSF (GM-CSF) and nitric oxide synthase (NOS). See, 

25 Klagsbrun, et al., Annu. Rev. Physiol., 53:217-239 (1991); Folkman, et al., J. Biol. 
Chem., 267:10931-10934 (1992) and Symes, et al.. Current Opinion in Lipidology, 
5:305-312 (1994). Muteins or fragments of an angiogenic growth factor may be used 
provided they induce nerve regeneration or alleviate a symptom of peripheral neuropathy. 
The feasibility of using recombinant formulations of angiogenic growth factors to 

30 expedite and/or augment collateral artery development in animal models of myocardial 
and hindlimb ischemia has been reported. See, Baffour, et al., supra (bFGF); Pu, et al.. 
Circulation, 88:208-215 (1993) (aFGF); Yanagisawa-Miwa, et al., supra (bFGF); Ferrara, 



wo 00/62798 



PCTAJS00/0976S 



-12- 

et al., Biochem. Biophys. Res. Commun., 161:851-855 (1989) (VEGF). In addition, 
therapeutic angiogenesis has been reported in the same or closely related models 
following administration of recombinant endothelial cell growth factor (ECGF) (Pu, et. 
al.. Circulation, 88:208-215 (1993)) and VEGF (Takeshita, et al.. Circulation, 90:228- 
5 234 (1994) supra). Previous studies, employing the animal model of chronic limb 
ischemia, reported an efficacy of intra-muscular endothelial cell growth factor (ECGF) 
(Pu, et al.. Circulation, 88:208-215 (1993) or VEGF (Takeshita, et al., Circulation, 
90:228-234 (1994) supra) administration. None of these references have suggested a role 
for angiogenic growth factors for treating peripheral neuropathy. 

10 VEGF is a particularly preferred angiogenic growth factor. Any of the VEGF 

isoforms (e.g., VEGF 1, 2, 3, 4, and 5) may be used in accordance v^th the methods of 
the invention. VEGF reportedly is an endothelial cell-specific mitogen (Ferrara, et al., 
Biochem Biophys Res Commun., 161 :85l-855, (1989), Keck, et al.. Science, 
246:1309-1312 (1989), and Plouet, et al., Embo J., 3801-3806 (1989)). VEGF was 

15 purified independently as a tumor-secreted factor that included vascular permeability by 
the Miles assay (Keck, et al., supra, and Connolly, et al., J. Biol. Chem., 
264:20017-20024 (1989)), and thus has an altemate designation, vascular permeability 
factor (VPF). Two features distinguish VEGF fi-om other heparin-binding, angiogenic 
growth factors. First, the NH2 terminus of VEGF is preceded by a typical signal 

20 sequence; therefore, unlike bFGF, VEGF can be secreted by intact cells. Second, its 
high-affinity binding sites, shown to include the tyrosine kinase receptors Flt-1 and 
Flt-1/KDR are present on endothelial cells. Ferrara, et al., supra, and Conn, et al., Proc. 
Natl. Acad. Sci. USA, 87:1323-1327 (1990). DNA encoding VEGF is disclosed in U.S. 
Pat. No. 5,332,671, the disclosure of which is herein incorporated by reference. 

25 Preferably, the angiogenic growth factor contains a secretory signal sequence that 

facilitates secretion of the protein. Angiogenic growth factors having native signal 
sequences, e.g., VEGF, are preferred. Angiogenic growth factors that do not have native 
signal sequences, e.g., bFGF, can be modified to contain such sequences using routine 
genetic manipulation techniques. See, Nabel, et al.. Nature, 362:844 (1993). 

30 The nucleotide sequence of numerous peptides and proteins, including the 

angiogenic growth factors, are readily available through a number of computer data 
bases, for example, GenBank, EMBL and Swiss-Prot. Using this information, a DNA or 



wo 00/62798 PCT/USOO/09765 

-13- 

RNA segment encoding the desired may be chemically synthesized or, alternatively, such 
a DNA or RNA segment may be obtained using routine procedures in the art, e.g, PGR 
amplification. 

To simplify the manipulation and handling of the nucleic acid encoding the 
5 growth factor, the nucleic acid preferably is inserted into a cassette where it is operably 
linked to a promoter. The promoter must be capable of driving expression of the mitogen 
in the desired target host cell, e.g., an endothelial cell, a muscle cell, a nerve cell. The 
selection of appropriate promoters can readily be accomplished. Preferably, a high 
expression promoter is used. An example of a suitable promoter is the 763 -base-pair 

10 cytomegalovirus (CMV) promoter. The Rous sarcoma virus (RSV) (Davis, et al.. Hum 
Gene Ther 4:151 (1 993)) and MMT promoters may also be used. Certain proteins can be 
expressed using their native promoter. Other elements that can enhance expression can 
also be included such as an enhancer or a system that results in high levels of expression 
such as a tat gene and tar element. This cassette can then be inserted into a vector, e.g., a 

1 5 plasmid vector such as pUCl 1 8, pBR322, or other known plasmid vectors, that includes, 
for example, an E. coli origin of replication. See, Sambrook, et al.. Molecular Cloning: A 
Laboratory Manual, Cold Spring Harbor Laboratory press, (1989). A plasmid vector may 
also include a selectable marker such as the P-lactamase gene for ampicillin resistance, 
provided that the marker polypeptide does not adversely effect the metabolism of the 

20 organism being treated. The cassette can also be bound to a nucleic acid binding moiety 
in a synthetic delivery system, such as the system disclosed in WO 95/122618. 

If desired, the DNA may also be used with a micro delivery vehicle such as 
cationic liposomes and adenoviral vectors. For a review of the procedures for liposome 
preparation, targeting and delivery of contents, see Mannino and Gould-Fogerite, Bio 

25 Techniques, 6:682 (1988). See also. Feigner and Holm, Bethesda Res. Lab. Focus, 1 1 
(2):21 (1989) and Maurer, R. A., Bethesda Res. Lab. Focus, 1 1(2):25 (1989). 

Replication-defective recombinant adenoviral vectors, can be produced in 
accordance with known techniques. See, Quantin, et al., Proc. Natl. Acad. Sci. USA, 
89:2581-2584 (1992); Stratford-Perricadet, et al., J. Clin. Invest., 90:626-630 (1992); and 

30 Rosenfeld, et al.. Cell, 68:143-155 (1992). 

In certEiin situations, it may be desirable to use nucleic acids encoding two or 
more different proteins in order to optimize the therapeutic outcome. For example, DNA 



wo 00/62798 PCTA;S00/a9765 

-14- 

encoding two angiogenic growth factors, e.g., VEGF and bFGF, can be used to provide 
an improved treatment over the use of bFGF alone. Alternatively, an angiogenic growth 
factor nucleic acid can be combined with other genes or their encoded gene products to 
enhance the activity of targeted cells, while simultaneously inducing angiogenesis, if 
5 desired, including, for example, nitric oxide synthase, L-arginine, fibronectin, urokinase, 
plasminogen activator and heparin. 

In order to facilitate injection, the nucleic acid is formulated with a 
pharmaceutically acceptable carrier. Examples of suitable carriers include, saline, 
albxmiin, dextrose and sterile water. The nucleic acid is injected into the ischemic tissue 

10 using standard injection techniques by use of, for example, a hypodermic needle. 

Hypodermic needle sizes from no. 29 to no. 16 are preferred. The nucleic acid may also 
be injected by an externally applied local injection apparatus, such as that used to inject 
antigens for allergy testing; or a transcutaneous "patch" capable of delivery to 
subcutaneous muscle. In general, the effective dose of the nucleic acid will be a function 

15 of the particular expressed protein, the target tissue, the patient and his or her clinical 
condition. Effective amounts of DNA typically are between about 1 and 4000 pg, more 
preferably from about 1000 to 4000 ^g and, most preferably, from about 2000 to 4000 

Once injected, the nucleic acid capable of expressing the desired angiogenic 
20 grov^ factor is taken up and expressed by the cells of the tissue. Because the vectors 
containing the nucleic acid of interest are not normally incorporated into the genome of 
the cells, expression of the protein of interest takes place for only a limited time. 
Typically, the angiogenic growth factor is expressed at therapeutic levels for about two 
days to several weeks, preferably for about 1-2 weeks. Reinjection of the DNA can be 
25 utilized to provide additional periods of expression of the angiogenic growth factor. If 
desired, a retrovirus vector can be used to incorporate the heterologous DNA into the 
genome of the cells and, thereby, increase the length of time during which the therapeutic 
polypeptide is expressed, from several weeks to indefinitely. 

The invention is not limited to treatment of ischemic tissue, but rather, is useful 
30 for treating peripheral neuropathies of various origin. Exemplary conditions that are 

characterized by peripheral neuropathy are known to those of ordinary skill in the art and 
include, but are not limited to, the following categories of disorders: (a) ischemic 



wo CO/62798 PCTAJSOO/09765 

-15- 

peripheral neuropathies; (b) toxin-induced peripheral neuropathies; (c) neuropathies 
associated with systemic disease; and (d) genetically acquired peripheral neuropathies. 

Exemplary ischemic peripheral neuropathies include neuropathies associated with 
ischemic tissues such as those associated with a diabetic condition, peripheral vascular 
5 disease, or other vascular insufficiency. 

Exemplary toxin-induced peripheral neuropathies are described in U.S. Patent No. 
5,648,335, entitled "Prevention and treatment of peripheral neuropathy", issued to Lewis, 
et al., and include neuropathies that are caused by neurotoxic agents including, 
therapeutic drugs, antineoplastic agents, contaminants in foods or medicinals, and 

10 environmental and industrial pollutants. By "toxic agent", or neurotoxic agent, is meant 
a substance that through its chemical action injures, impairs, or inhibits the activity of a 
component of the nervous system. The list of neurotoxic agents that cause neuropathies is 
lengthy, and includes, but is not limited to, neoplastic agents such as vincristine, 
vinblastine, cisplatin, taxol, or dideoxy-compounds, e.g., dideoxyinosine; alcohol; 

15 metals; industrial toxins involved in occupational or environmental exposure; 

contaminants of food or medicinals; or over-doses of vitamins or therapeutic drugs, e.g., 
antibiotics such as penicillin or chloramphenicol, or megadoses of vitamins A, D, or B^. 
An extensive, although not complete, list of chemical compounds with neurotoxic 
side-effects is found in Table 1 . Although this list provides examples of neurotoxic 

20 compoimds, it is intended to exemplify, not limit, the scope of the invention. Other toxic 
agents can cause neuropathies, and can be characterized by methods known to one skilled 
in the art. By "exposure to a toxic agent" is meant that the toxic agent is made available 
to, or comes into contact v^th, a mammal of the invention. Exposure to a toxic agent can 
occur by direct administration, e.g., by ingestion or administration of a food, medicinal, 

25 or therapeutic agent, e.g., a chemotherapeutic agent, by accidental contamination, or by 
environmental exposure, e.g., aerial or aqueous exposure. 



TABLE 1: AGENTS THAT CAUSE PERIPHERAL NEUROPATHY 


AGENT 


ACTIVITY 


Acetazolamide 


diuretic 


Acrylimide 


flocculent, grouting agent 


Adriamycin 


antineoplastic 


alcohol (ethanol) solvent 


recreational drug 



wo 00/62798 



-16- 



PCT/USOO/09765 



Almitrine 


resniratorv stimulant 


Amiodarone 


antiarrhythmic 


Amnhotericin 


antimicrobial 


Ar^jPTiic 


herbicide insecticide 


Aurothioel uco se 


antirheumati c 


B arbi turates 


anticonvulsant sedative 


Buckthorn 


toxic berry 


Carbamates 


insecticide 


carbon disulfide f CS-^"! 


industrial 


chloramnhenicol 


antibacterial 


phlnrnniiirip 


antimalarial 

CUllililAlCU iCll 


f*li n 1 6* <? tvra tn i n e 


antih vnerl innnrntf^i nf*m i c 




antinennla^tic 


rlinniiinnl ampbipide 


antibacterial 


rnlp<!tinnl 


an ti h vnerl i nnnrntpi n pm t c 


colchicine 


20ut sunnressant 


rnli<!tin 


antimicrobial 


rvcln^prinp 


antibacterial 


cvtarahine 


antineonlastic 


dansone 


dermatolosic including lenrosv 

wWA XAAU-lV/XV/^A W AAA WA U.U.AAAK AWL/X T 


dideoxvcvtidine 


antineonlastic 

UX X U AX W L/ A UiJ VX W 


dideoxvinosine 

\iXXUWV/V T UXx/iSUxW 


antine on 1 asti c 


d 1 d en wth vm 1 d i Ti e 


antiviral 


disulfiram 


anti alcohol 

WXX^XUAW\/A Av A 


dftYnnihif^in 


antinpnnla<3tip 

CU A Li i 1& V/ LI 1 do Li W 


pthamhiitol 

wUiCUiiUULVfi 


antibartprial 


ethionamide 


antibacterial 


giutethimide 


sedative, hypnotic 


gold 


antirheumatic 


hexacarbons solvents 


hormonal contraceptives 


hexamethylolmelamine 


fireproofing, creaseproofing 



wo 00/62798 



-17- 



PCTAJS0O/Oy76S 



hydralazine 


antihypertensive 


hydroxychloroquine 


antirheumatic 


imipramine 


antidepressant 


indomethacin 


anti-inflammatory 


inorganic lead 


toxic metal in paint, etc. 


isoniazid 


antituberculous 


lithiiim 


antidepressant 


methylmercury 


industrial waste 


metformin 


antidiabetic 


methylhydrazine 


synthetic intermediate 


metronidazole 


antiprotozoal 


misonidazole 


radiosensitizer 


nitrofurantoin 


urinary antiseptic 


nitrogen mustard antineoplastic 


nerve gas 


nitrous oxide 


anesthetic 


organophosphates 


insecticides 


ospolot 


anticonvulsant 


penicillin 


antibacterial 


perhexiline 


antiarrhythmic 


perhexiline maleate 


antianhythmic 


phenytoin 


anticonvulsant 


platinum 


drug component 


primidone 


anticonvulsant 


procarbazine 


antineoplastic 


pyridoxine 


vitamin B6 


sodium cvanate 


anti-sickling 


streptomycin 


antimicrobial 


sulphonamides 


antimicrobial 


suramin 


antineoplastic 


tamoxifen 


antineoplastic 


taxol 


antineoplastic 



wo 00/62798 



-18- 



PCT/USOO/09765 



thnltdftiniHe 


antileorous 


thalliiiTn 
uxcuix mil 


rat poison 


tri a m t eren e 


diuretic 


trimpfVivltin 

U JlllCUIjrlliil 


tnyir metal 




health food additive 


vincristine 


antineoplastic 


vinblastine 


antineoplastic 


vindesine 


antineoplastic 


vitamin A 


mega doses 


vitamin D 


mega doses 



In general, neurotoxicity is dose-related, and presents as reduced intestinal 
motility and peripheral neuropathy, especially in the distal muscles of the hands and feet, 
postural hypotension, and atony of the urinary bladder. Similar problems have been 

5 reported with taxol and cisplatin (Mollman, J. E., 1990, New Eng Jour Med. 

322:126-127), although cisplatin-related neurotoxicity reportedly can be alleviated with 
nerve growth factor (NGF) (Apfel, S. C, et al, 1992, Annals of Neurology 3 1 :76-80). 
Although the neurotoxicity is sometimes reversible after removal of the neurotoxic agent, 
recovery reportedly can be a very slow process (Legha, S., 1986, Medical Toxicology 

10 1:421-427; Olesen, et al., 1991, Drug Safety 6:302-314). 

Exemplary neuropathies associated with a systemic condition include: uremia, 
childhood cholestatic liver disease, chronic respiratory insufficiency, alcoholic 
polyneuropathy, multiple organ failure, sepsis, hypoalbuminemia, eosinophilia-myalgia 
syndrome, hepatitis, porphyria, hypoglycemia, vitamin deficiency, chronic liver disease, 

15 primary biliary cirrhosis, hyperlipidemia, leprosy, Lyme disease, herpes zoster, 

Guillain-Barre syndrome, chronic inflanunatory demyelinating polyradiculoneuropathy, 
sensory perineuritis, acquired immunodeficiency syndrome (AIDS)-associated 
neuropathy, Sjogren's syndrome, primary vasculitis (such as polyarteritis nodosa), 
allergic granulomatous angiitis (Churg-Strauss), hypersensitivity angiitis, Wegener's 

20 granulomatosis, rheumatoid arthritis, systemic lupus erythematosis, mixed connective 
tissue disease, scleroderma, sarcoidosis, vasculitis, systemic vasculitides, acute 
inflammatory demyelinating polyneuropathy, post-polio syndrome, carpal tunnel 



wo 00/62798 



.19- 



PCT/US00/0976S 



syndrome, pandysautonomia, primaiy systemic amyloidosis, hypothyroidism, chronic 
obstructive pulmonary disease, acromegaly, malabsorption (sprue, celiac disease), 
carcinomas (sensory, sensorimotor, late and demyelinating), lymphoma (including 
Hodgkin's), polycythemia vera, multiple myeloma (lytic type, osteosclerotic, or solitary 
5 plasmacytoma), benign monoclonal gammopathy, macroglobulinemia, and 
cryoglobulinemia. 

Exemplary genetically acquired neuropathies include: peroneal muscular atrophy 
(Charcot-Marie-Tooth Disease, types I, II, and X), hereditary amyloid neuropathies, 
hereditary sensory neuropathy (type I and type II), porphyric neuropathy, hereditary 

10 liability to pressure palsy, Fabry's disease, adrenomyeloneuropathy, Riley-Day 
syndrome, Dejerine-Sottas neuropathy (hereditary motor-sensory neuropathy-III), 
Refsum's disease, ataxia-telangiectasia, hereditary tyrosinemia, anaphalipoproteinemia, 
abetalipoproteinemia, giant axonal neuropathy, metachromatic leukodystrophy, globoid 
cell leukodystrophy, and Friedrich's ataxia. Also included in the invention are 

15 mononeuropathy multiplex, plexopathy, and pure motor neuropathy. 

The angiogenic growth factors of the invention are administered in effective 
amoxmts. An effective amount is a dosage of the angiogenic growth factor nucleic acid 
sufficient to provide a medically desirable result. The effective amount will vary with the 
particular condition being treated, the age and physical condition of the subject being 

20 treated, the severity of the condition, the duration of the treatment, the nature of the 

concurrent therapy (if any), the specific route of administration and like factors within the 
knowledge and expertise of the healthcare practitioner. For example, in connection with 
peripheral neuropathy, an effective amount is that amoimt which alleviates a symptom of 
the neuropathy. Likewise, an effective amount for treating a subject who has sustained a 

25 peripheral nerve injury, would be an amount sufficient to enhance peripheral nerve 
regeneration. Thus, it will be understood that the angiogenic growth factor of the 
invention can be used to treat the above-noted conditions prophylactically in subjects at 
risk of developing the foregoing conditions. As used herein, "treat" embraces all of the 
foregoing. It is preferred generally that a maximum dose be used, that is, the highest safe 

30 dose according to sound medical judgment. 

A particularly important aspect of the invention involves the use of the angiogenic 
growth factors of the invention for treating subjects who have sustained a peripheral 



wo 00/62798 PCT/US00/d9765 

-20- 

neuropathy as a side effect of ischemic heart disease. Ischemia refers to a lack of oxygen 
due to inadequate perfusion of blood. Ischemic heart disease is characterized by a 
disturbance in cardiac function due to an inadequate supply of oxygen to the heart. The 
most common form of this disease involves a reduction in the lumen of coronary arteries, 
5 which limits coronary blood-flow. 

When ischemic heart disease becomes very serious, management of the disease 
becomes invasive. Until recently, ischemic heart disease was treated by coronary-artery, 
bypass surgery. Less invasive procedures, however, now have been developed. These 
procedures involve the use of catheters introduced into the narrowed region of the blood 

10 vessel ("the stenosis") for mechanically disrupting, laser ablating or dilating the stenosis. 
The most widely used method to achieve revascularization of a coronary artery is 
percutaneous transluminal coronary angioplasty. A flexible guide wire is advanced into a 
coronary artery and positioned across the stenosis. A balloon catheter then is advanced 
over the guide wire until the balloon is positioned across the stenosis. The balloon then is 

15 repeatedly inflated until the stenosis is substantially eliminated. This procedure, as 

compared to heart surgery, is relatively noninvasive and typically involves a hospital stay 
of only a few days. The procedure is an important tool in the management of serious 
heart conditions and can also be used to deliver the angiogenic growth factor of the 
invention to a local site of ischemic tissue and for treatment of a neuropathy in the 

20 ischemic tissue. Alternatively, the angiogenic growth factors can be intramuscularly 

injected directly into the ischemic tissue. In certain embodiments, the angiogenic growth 
factors are in the form of controlled release preparations for the sustained delivery of the 
factors to the ischemic or other tissue that presents symptoms of a peripheral neuropathy. 
Controlled release systems for delivery of an angiogenic growth factor nucleic acid or 

25 polypeptide are described in more detail below. 

A subject, as used herein, refers to any mammal (preferably, a human) that may 
be susceptible to a condition associated with peripheral neuropathy (such as the 
conditions described above). In certain embodiments, the manunal is otherwise free of 
symptoms calling for angiogenic growth factor treatment. Different aspects of the 

30 invention may exclude one or more of the following subject populations that present with 
a peripheral neuropathy: (1) patients presenting with a vascular disease; (2) patients 
presenting with a vascular obstruction (large vessels); (3) patients presenting with a 



wo 00/62798 PCT/US00/0976S 

-21- 

microvascular disease; (4) patients presenting with an ischemic tissue, such as an 
ischemic limb; (5) patients being treated with an angiogenic growth factor to promote 
revascularization; and (6) patients who are being treated using gene therapy. 

In some particular embodiments, the preferred vehicle is a biocompatible micro 
5 particle or implant that is suitable for implantation into the subject. Exemplary 

bioerodible implants that are useful in accordance with this method are described in PCT 
International application no. PCTAJS/03307 (Publication No. WO 95/24929, entitled 
"Polymeric Gene Delivery System", claiming priority to U.S. patent application serial no. 
213,668, filed March 15, 1994). PCT/US/0307 describes a biocompatible, preferably 

10 biodegradable polymeric matrix for containing an exogenous gene under the control of an 
appropriate promotor. The polymeric matrix is used to achieve sustained release of the 
exogenous gene in the subject. In accordance with the instant invention, the angiogenic 
growth factor nucleic acids described herein are encapsulated or dispersed within the 
biocompatible, preferably biodegradable polymeric matrix disclosed in PCT/US/03307. 

15 The polymeric matrix preferably is in the form of a micro particle such as a micro sphere 
(wherein the angiogenic growth factor nucleic acid is dispersed throughout a solid 
polymeric matrix) or a microcapsule (wherein the angiogenic growth factor nucleic acid 
is stored in the core of a polymeric shell). Other forms of the polymeric matrix for 
containing the angiogenic growth factor nucleic acid include films, coatings, gels, 

20 implants, and stents. The size and composition of the polymeric matrix device is selected 
to result in favorable release kinetics in the tissue into which the matrix device is 
implanted. The size of the polymeric matrix devise further is selected according to the 
method of delivery which is to be used, typically injection into a tissue or administration 
of a suspension by aerosol into the nasal and/or pulmonary areas. The polymeric matrix 

25 composition can be selected to have both favorable degradation rates and also to be 

formed of a material which is bioadhesive, to further increase the effectiveness of transfer 
when the devise is administered to a vascular surface. The matrix composition also can 
be selected not to degrade, but rather, to release by diffusion over an extended period of 
time. 

30 Both non-biodegradable and biodegradable polymeric matrices can be used to 

deliver the angiogenic growth factor nucleic acids of the invention to the subject. 
Biodegradable matrices are preferred. Such polymers may be natural or synthetic 



wo 00/62798 



-22- 



PCT/USOO/09765 



polymers. Synthetic polymers are preferred. The polymer is selected based on the period 
of time over which release is desired, generally in the order of a few hours to a year or 
longer. Typically, release over a period rangmg from between a few hours and three to 
twelve months is most desirable. The polymer optionally is in the form of a hydrogel that 
can absorb up to about 90% of its weight in water and further, optionally is cross-linked 
with multi-valent ions or other polymers. 

In general, the angiogenic growth factor nucleic acids of the invention are 
delivered using the bioerodible implant by way of diffusion, or more preferably, by 
degradation of the polymeric matrix. Exemplary synthetic polymers which can be used to 
form the biodegradable delivery system include: polyamides, polycarbonates, 
polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terepthalates, 
polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, poly-vinyl halides, 
polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and co-polymers 
thereof, alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro 
celluloses, polymers of acrylic and methacrylic esters, methyl cellulose, ethyl cellulose, 
hydroxypropyl cellulose, hydroxy-propyl methyl cellulose, hydroxybutyl methyl 
cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose 
acetate phthalate, carboxylethyl cellulose, cellulose triacetate, cellulose sulphate sodium 
salt, poly(methyl methacrylate), poly(ethyl methacrylate), poly(butylmethacrylate), 
poly(isobutyl methacrylate), poly(hexyhnethacrylate), poly(isodecyl methacrylate), 
poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), 
poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate), polyethylene, 
polypropylene, poly(ethylene glycol), poly(ethylene oxide), poly(ethylene terephthalate), 
poly(vinyl alcohols), polyvinyl acetate, poly vinyl chloride, polystyrene and 
polyvinylpyrrolidone. 

Examples of non-biodegradable polymers include ethylene vinyl acetate, 
poly(meth)acrylic acid, polyamides, copolymers and mixtures thereof. 

Examples of biodegradable polymers include synthetic polymers such as 
polymers of lactic acid and glycolic acid, polyanhydrides, poly(ortho)esters, 
polyurethanes, poly(butic acid), poly(valeric acid), and poly(lactide-cocaprolactone), and 
natural polymers such as alginate and other polysaccharides including dextran and 
cellulose, collagen, chemical derivatives thereof (substitutions, additions of chemical 



wo 00/62798 



-23- 



PCT/USOO/09765 



groups, for example, alkyl, alkylene, hydroxylations, oxidations, and other modifications 
routinely made by those skilled in the art), albumin and other hydrophilic proteins, zein 
and other prolamines and hydrophobic proteins, copolymers and mixtures thereof. In 
general, these materials degrade either by enzymatic hydrolysis or exposure to water in 

5 vivo, by surface or bulk erosion. 

Bioadhesive polymers of particular interest include bioerodible hydrogels 
described by H.S. Sawhney, CP. Pathak and J.A. Hubell in Macromolecules, 1993, 26, 
581-587, the teachings of which are incorporated herein, poly hyaluronic acids, casein, 
gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl 

10 methacrylates), poly(ethyl methacrylates), poly(butylmethacrylate), poly(isobutyl 
methacrylate), poly(hexylmethacrylate), poly(isodecyl methacrylate), poly(lauryl 
methacrylate), poly(phenyl methacrylate), poly(methyl acryiate), poly(isopropyl 
acrylate), poly(isobutyl acryiate), and poly(octadecyl acryiate). Thus, the invention 
provides a composition of the above-described angiogenic growth factors for use as a 

15 medicament, methods for preparing the medicament and methods for the sustained 

release of the medicament in vivo. In the preferred embodiments, the angiogenic growth 
factor nucleic acid is a human VEGF nucleic acid, alone or in combination v^th a human 
bFGF nucleic acid. Preferably, the angiogenic growth factor nucleic acid is operably 
linked to a gene expression sequence to permit expression of the angiogenic growth 

20 factor nucleic acid in the target cell. 

Compaction agents also can be used alone, or in combination with, a vector of the 
invention. A "compaction agent", as used herein, refers to an agent, such as a histone, 
that neutralizes the negative charges on the nucleic acid and thereby permits compaction 
of the nucleic acid into a fine granule. Compaction of the nucleic acid facilitates the 

25 uptake of the nucleic acid by the target cell. The compaction agents can be used alone, 
i.e., to deliver the isolated angiogenic growth factor nucleic acid in a form that is more 
efficiently taken up by the cell or, more preferably, in combination with one or more of 
the above-described vectors. 

Other exemplary compositions that can be used to facilitate uptake by a target cell 

30 of the angiogenic growth factor nucleic acids include calcium phosphate and other 
chemical mediators of intracellular transport, microinjection compositions, 
electroporation and homologous recombination compositions (e.g., for integrating an 



wo 00/62798 



-24- 



PCT/USOO/09765 



angiogenic growth factor nucleic acid into a preselected location within the target cell 
chromosome). 

The angiogenic growth factor nucleic acids code for an angiogenic growth factor 
polypeptide. As used herein, a "angiogenic growth factor polypeptide" refers to a 
5 polypeptide that, either directly or indirectly, enhances endothelial cell growth. 

Angiogenic growth factor polypeptides are useful for alleviating a symptom of peripheral 
neuropathy and/or enhancing nerve regeneration. The preferred angiogenic growth factor 
polypeptides of the invention are the human VEGF isoforms, administered alone or in 
combination with human bFGF. Angiogenic growth factor polypeptides further embrace 

10 functionally equivalent variants, and analogs of angiogenic growth factors, provided that 
the fragments, variants, and analogs alleviate a symptom of a peripheral neuropathy 
and/or enhance nerve regeneration. The invention also embraces proteins and peptides 
coded for by any of the foregoing angiogenic growth factor nucleic acids. The invention 
also embraces agents that upregulate expression of an angiogenic growth factor 

1 5 polypeptide in vivo . 

A "functionally equivalent variant" of an angiogenic growth factor is capable of 
alleviating a symptom of a peripheral neuropathy and/or enhancing nerve regeneration in 
vitro or in vivo. An in vitro assay or an in vivo animal model (see, e.g., the Examples) 
can be used as a screening assay to measure the ability of a polypeptide to alleviate a 

20 symptom of a peripheral neuropathy and/or enhance nerve regeneration. The animal 
model disclosed in the Examples can be used to screen therapeutic drugs because it is 
predictive of the ability of the polypeptide to treat a peripheral neuropathy in vivo. 
Exemplary "functionally equivalent variants" of the angiogenic growth factors (such as 
the exemplary growth factors disclosed herein) include fragments of these factors, as well 

25 as polypeptide analogs of these factors which contain conservative amino acid 

substitutions, provided that the polypeptide variants and analogs are capable of alleviating 
a symptom of a peripheral neuropathy and/or enhancing nerve regeneration. 

It will be appreciated by those skilled in the art that various modifications of the 
angiogenic growth factor polypeptide having the sequences deposited in the publicly 

30 available databases or functionally equivalent fi-agments thereof can be made v^dthout 
departing from the essential nature of the invention. Accordingly, it is intended that 
polypeptides which have the published amino acid sequences but which include 



wo 00/62798 PCT/USOO/09765 

-25- 

conservative substitutions are embraced within the instant invention. As used herein, 
"conservative amino acid substitution" refers to an amino acid substitution which does 
not alter the relative charge or size characteristics of the polypeptide in which the amino 
acid substitution is made. Conservative substitutions of amino acids include substitutions 
5 made amongst amino acids with the foUov^ng groups: (1) M,I,L,V; (2) F,Y,W; (3) 
K,R,H; (4) A,G; (5) S,T; (6) Q,N; and, (7) E,D. Fusion proteins, in which a peptide of 
the invention is coupled to a solid support (such as a polymeric bead for controlled 
release), or a reporter group (such as radiolabel or other tag), also are embraced within 
the invention. 

10 When used therapeutically, the isolated angiogenic growth factors of the invention 

are administered in therapeutically effective amounts. In general, a therapeutically 
effective amount means that amount necessary to delay the onset of, inhibit the 
progression of, or halt altogether the particular condition being treated. As noted above, a 
therapeutically effective amount will vary with the subject's age, condition, and sex, as 

15 well as the nature and extent of the disease in the subject, all of which can be determined 
by one of ordinary skill in the art. The dosage may be adjusted by the individual 
physician or veterinarian, particularly in the event of any complication. A therapeutically 
effective amoimt can vary throughout a broad dosage range, e.g., from 0,01 mg/kg to 
about 1000 mg/kg, preferably from about 0.1 mg/kg to about 200 mg/kg, and most 

20 preferably from about 0.2 mg/kg to about 20 mg/kg, in one or more dose administrations 
daily, for one or more days. In certain aspects of the invention described above, the 
angiogenic growth factors are administered over a period of months to years in the case 
of a chronic peripheral neuropathy. 

The therapeutically effective amount of the isolated angiogenic growth factor is 

25 that amount effective to inhibit the development of peripheral neuropathy, alleviate a 
symptom of a peripheral neuropathy, and/or enhance nerve regeneration as determined 
by, for example, standard tests known in the art. It is believed that the angiogenic grov^ 
factors directly and/or indirectly enhance nerve regeneration in the vicinity of the target 
cell. Diagnostic tests that are used to diagnose a peripheral neuropathy can by used to 

30 select an effective amount of the angiogenic growth factor. In vitro assays are available 
to determine whether a factor has been effective in inducing nerve regeneration. 



wo 00/62798 



.26- 



PCTAJS00/0976S 



Optionally, the isolated angiogenic growth factor is administered to the subject in 
combination with an alternative method for treating the neuropathy or for treating the 
particular condition that is associated with the peripheral neuropathy. See, e.g., 
Harrisons, Principles of Internal Medicine (McGraw Hill, Inc., New York) for a 
5 description of standard treatments for peripheral neuropathies and/or for conditions that 
also present with symptoms of a peripheral neuropathy. The method for treating 
neuropathy may be a surgical method, a drug for treating neuropathy, a gene therapy 
method or a combination of the foregoing. 

Surgical methods for treating a condition of vascular insufficiency include 

10 procedures such as bypass surgery, atherectomy, laser procedures, ultrasonic procedures, 
and balloon angioplasty to enhance vascularization of an ischemic tissue. In certain 
embodiments, the isolated angiogenic growth factor is administered to a subject in 
combination with a balloon angioplasty procedure. The isolated angiogenic grov^ factor 
is attached to the balloon angioplasty catheter in a manner which permits release of the 

15 isolated angiogenic growth factor at the site of the atherosclerotic plaque. The isolated 
angiogenic growth factor may be attached to the balloon angioplasty catheter in 
accordance with standard procedures known in the art. See, e.g., U.S. Patent No. 
5,652,225, entitled "Methods and products for nucleic acid delivery", issued to J. Isner, 
for a description of a balloon angioplasty procedure for delivering VEGF. 

20 Additionally, the angiogenic growth factor may be administered in combination 

with the toxic agent which causes the neuropathy, e.g., a neoplastic agent, to alleviate the 
symptoms of peripheral neuropathy that are a side effect of the neoplastic agent. 

The angiogenic growth factor also may be administered in combination with a 
drug for treating the condition which is believed to be associated with, or caused by, the 

25 peripheral neuropathy. For example, the angiogenic growth factor may be administered 
in combination with a drug for treating a diabetic condition (e.g., insulin), to alleviate the 
symptoms of peripheral neuropathy that are a side effect of the diabetic condition. 

The above-described drug therapies are well known to those of ordinary skill in 
the art and are administered by modes known to those of skill in the art. The drug 

30 therapies are administered in amounts which are effective to achieve the physiological 
goals (to prevent or reduce the physiological consequences of a peripheral neuropathy), in 
combination with the isolated angiogenic growth factor(s) of the invention. Thus, it is 



wo 00/62798 PCT/US00/0y765 

-27- 

contemplated that the drug therapies may be administered in amounts which are not 
capable of preventing or reducing the physiological consequences of the peripheral 
neuropathy when the drug therapies are administered alone but which are capable of 
preventing or reducing the physiological consequences of the peripheral neuropathy when 
5 administered in combination with the isolated angiogenic growth factors of the invention. 
The isolated angiogenic growth factor may be administered alone or in 
combination with the above-described drug therapies as part of a pharmaceutical 
composition. Such a pharmaceutical composition may include the isolated angiogenic 
growth factor in combination with any standard physiologically and/or pharmaceutically 

10 acceptable carriers which are known in the art. The compositions should be sterile and 
contain a therapeutically effective amount of the isolated angiogenic growth factor in a 
unit of weight or volume suitable for administration to a patient. The term 
"pharmaceutically-acceptable carrier" as used herein means one or more compatible solid 
or liquid filler, diluents or encapsulating substances which are suitable for administration 

15 into a human or other animal. The tenn "carrier" denotes an organic or inorganic 

ingredient, natural or synthetic, with which the active ingredient is combined to facilitate 
the application. The components of the pharmaceutical compositions also are capable of 
being co-mingled with the molecules of the present invention, and with each other, in a 
manner such that there is no interaction which would substantially impair the desired 

20 pharmaceutical efficacy. Pharmaceutically acceptable fiirther means a non-toxic material 
that is compatible vrfth a biological system such as a cell, cell culture, tissue, or organism. 
The characteristics of the carrier will depend on the route of administration. 
Physiologically and pharmaceutically acceptable carriers include diluents, fillers, salts, 
buffers, stabilizers, solubilizers, and other materials which are well known in the art. 

25 Compositions suitable for parenteral administration conveniently comprise a 

sterile aqueous preparation of the angiogenic growth factors, which is preferably isotonic 
with the blood of the recipient. This aqueous preparation may be formulated according to 
known methods using suitable dispersing or wetting agents and suspending agents. The 
sterile injectable preparation also may be a sterile injectable solution or suspension in a 

30 non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3- 
butane did. Among the acceptable vehicles and solvents that may be employed are 
water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed 



wo 00/62798 PCT/USOO/09765 

-28- 

oils are conventionally employed as a solvent or suspending medium. For this purpose, 
any bland fixed oil may be employed including synthetic mono- or di-glycerides. In 
addition, fatty acids such as oleic acid may be used in the preparation of injectables. 
Carrier formulations suitable for oral, subcutaneous, intravenous, intramuscular, etc. 
5 administrations can be foimd in Remington's Pharmaceutical Sciences, Mack Publishing 
Co., Easton, PA. 

A variety of administration routes are available. The particular mode selected will 
depend, of course, upon the particular drug selected, the severity of the condition being 
treated, and the dosage required for therapeutic efficacy. The methods of the invention, 

10 generally speaking, may be practiced using any mode of administration that is medically 
acceptable, meaning any mode that produces effective levels of the active compoimds 
without causing clinically unacceptable adverse effects. Intramuscular administration is 
preferred; however other modes of administration may be acceptable including, e.g., 
other parenteral routes, oral, rectal, topical, nasal, or interdermal. The term "parenteral" 

15 includes subcutaneous, intravenous, intramuscular, or infusion. 

The pharmaceutical compositions may conveniently be presented in unit dosage 
form and may be prepared by any of the methods well-known in the art of pharmacy. All 
methods include the step of bringing the angiogenic growth factors into association with a 
carrier which constitutes one or more accessory ingredients. In general, the compositions 

20 are prepared by uniformly and intimately bringing the angiogenic growth factors into 
association with a liquid carrier, a finely divided solid carrier, or both, and then, if 
necessary, shaping the product. 

Other delivery systems can include time-release, delayed release or sustained 
release delivery systems such as those described in detail above. In general, such systems 

25 can avoid repeated administrations of the angiogenic growth factors, increasing 

convenience to the subject and the physician. Many types of release delivery systems are 
available and known to those of ordinary skill in the art. They include the above- 
described polymeric systems, as well as polymer base systems such as poly(lactide- 
glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, 

30 polyhydroxybutyric acid, and polyanhydrides. Microcapsules of the foregoing polymers 
containing drugs are described in, for example, U.S. Patent 5,075,109. Delivery systems 
also include non-polymer systems that are: lipids including sterols such as cholesterol. 



wo 00/62798 PCTAJSOO/09765 

-29- 

cholesterol esters and fatty acids or neutral fats such as mono- di- and tri-glycerides; 
hydrogel release systems; sylastic systems; peptide based systems; wax coatings; 
compressed tablets using conventional binders and excipients; partially fused implants; 
and the like. Specific examples include, but are not limited to: (a) erosional systems in 

5 which the angiogenic growth factor is contained in a form within a matrix such as those 
described in U.S. Patent Nos. 4,452,775, 4,667,014, 4,748,034 and 5,239,660 and (b) 
diffiisional systems in which an active component permeates at a controlled rate from a 
polymer such as described in U.S. Patent Nos. 3,832,253, and 3,854,480. In addition, 
pump-based hardware delivery systems can be used, some of which are adapted for 

10 implantation. 

Use of a long-term sustained release implant may be particularly suitable for 
treatment of chronic conditions. Long-term release, are used herein, means that the 
implant is constructed and arranged to delivery therapeutic levels of the active ingredient 
for at least 30 days, and preferably 60 days. Long-term sustained release implants are 
15 well-known to those of ordinary skill in the art and include some of the release systems 
described above. 

The isolated angiogenic growth factor may be administered alone or in 
combination with the above-described drug therapies by any conventional route, 
including injection or by gradual infusion over time. The administration may, for 

20 example, be intramuscular, intravenous, intraperitoneal, intra-cavity, subcutaneous, oral, 
or transdermal. When using the isolated angiogenic growth factor of the invention, direct 
administration to the nerve injury site, such as by administration in conjunction with a 
balloon angioplasty catheter or intramuscular injection, is preferred. 

Preparations for parenteral administration include sterile aqueous or non-aqueous 

25 solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene 
glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters 
such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, 
emulsions or suspensions, including saline and buffered media. Parenteral vehicles 
include sodium chloride solution. Ringer's dextrose, dextrose and sodium chloride, 

30 lactated Ringer's or fixed oils. Intravenous vehicles include fluid and nutrient 

replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the 



wo 00/62798 PCT/USOO/09765 

-30- 

like. Preservatives and other additives may also be present such as, for example, 
antimicrobials, anti-oxidants, chelating agents, and inert gases and the like. 

In general, the angiogenic grovslh factor nucleic acids can be administered to the 
subject (any mammalian recipient) using the same modes of administration that currently 
5 are used for gene therapy in humans (e.g., adenovirus-mediated gene therapy). 

Preferably, the angiogenic growth factor nucleic acid (contained in, or associated wdth, an 
appropriate vector) is administered to the mammalian recipient by balloon angioplasty 
catheter (described above), or intra-vascular, or, more preferably, intramuscular injection. 
A procedure for performing in vivo gene therapy for delivering a nucleic acid encoding 
10 an intact angiogenic grov^ factor (VEGF) to cells in vivo for treating a vascular injury is 
reported in U.S. Patent No. 5,830,879, entitled "Treatment of vascular injury using 
vascular endothelial growth factor", issued to J. Isner. 

As an illustrative example, a vector containing a VEGF nucleic acid is delivered 
to a site of vascular injury in a subject who is a candidate for such gene therapy. Then, 
15 the vector genetically modifies the target cells (e.g., endothelial cells, muscle cells, nerve 
cells) in vivo v^th DNA (RNA) encoding the VEGF. Such genetically modified cells 
express VEGF which is believed to directly or indirectly alleviate the symptoms of a 
peripheral neuropathy in vivo. Although not wishing to be bound to a particular theory 
or mechanism, it is believed that such genetically modified cells exhibit enhanced nerve 
20 regeneration in vitro and in vivo. 

The invention will be more fully understood by reference to the following 
examples. These examples, however, are merely intended to illustrate the embodiments 
of the invention and are not to be construed to limit the scope of the invention. 

EXAMPLES 

25 Introduction: 

The Examples employ certain procedures which are known in the art and which 
may have been published in issued U.S. patents or foreign patent documents, including 
the following documents: 



Patent/Serial/Publication 
No. 


Title 


Inventor 


U.S. 5,830,879 


Treatment of vascular injury 
using vascular endothelial 
growth factor 


J. Isner 



wo 00/62798 PCTAJSOO/09765 

-31- 



U.S. 5,652,225 


Methods and products for 
nucleic acid delivery 


J. Isner 


U.S. Serial No. 08/545,998 


Method for Treating 
Ischemic Tissue 


J. Isner 


U.S. 5,648,335; 
U.S. 5,633,228; and 
U.S. 5,569,648 


Prevention and treatment of 
peripheral neiu-opathy 


M. Lewis, et al. 



Selected methods that previously have been described in the above-identified 
patent documents and that can be used in accordance with the methods of the instant 
invention are briefly summarized herein. 

5 Method; Plasmids 

Complementary DNA clones for recombinant human VEFG]65 isolated from 
cDNA libraries prepared from HL60 leukemia cells, were assembled into a simple 
eukaryotic expression plasmid that utilizes 736 bp cytomegalovirus promoter/enhancer to 
drive VEGF expression. Downstream from the VEGF cDNA is an SV40 polyadenylation 

10 sequence. Also included in this plasmid is a fragment containing the SV40 origin of 
replication that includes the 72 bp repeat, but this sequence is not fiinctionally relevant 
(for autonomous replication) in the absence of SV40 T antigen. These fragments occur in 
the pUCl 1 8 vector which includes an E. Coli origin of replication and the P-galactosidase 
gene for ampicillin resistance. The biological activity of VEGF165 secreted from cells 

15 transfected wdth this construct (phVEGFies) was previously confirmed by evidence that 
media conditioned by transfected human 293 cells promoted the proliferation of capillary 
cells (Leung, et al. Science, 246:1306-9 (1989)). 

The plasmid pGSVLacZ (courtesy of Dr. Claire Bonnerot) containing a nuclear 
targeted P-galactosidase sequence coupled to the simian virus 40 early promoters 

20 (Bonnerot, et al., Proa Natl AcadSci, US.A., 84:6795-9 (1987)) was used for all the 
control transfections. 
Method: Animal Model 

New Zealand white rabbits with operatively induced unilateral hindlimb vascular 
insufficiency, (Takeshita, et al., Circulation, 90:228-234 (1994); Takeshita, et al., J, Clin, 

IS Invest, 93:662-70 (1994); Pu, et al.. Circulation, 88:208-215 (1993), were used to model 
both acute and chronic ischemia. All protocols were approved by the Institutional 
Animal Care and Use Committee. The care of animals complied with the guidelines of 



wo 00/62798 



"32- 



PCTAJSOO/09765 



the Canadian Council of Animal Care, the Principles of Laboratory Animal Care, and the 
Guide for the Care and Use of Laboratory Animals (NIH publication No. 80-23, revised 
1985). Fifty-nine male New Zealand White rabbits (mean weight = 3 kg) were 
anesthetized with ketamine (50 mg/kg) and xylazine (5 mg/kg). Through a longitudinal 

5 incision performed in a medial thigh, the femoral artery was dissected free along its entire 
length, as were all major branches of the femoral artery, including the inferior epigastric, 
deep femoral, lateral circumflex and superficial epigastric arteries. After ftirther 
dissecting the popliteal and saphenous arteries distally, the external iliac artery as well as 
all of the above arteries were ligated. Finally, the femoral artery was completely excised 

10 from its proximal origin as a branch of the external iliac artery to the point distally where 
it bifiarcates into the saphenous and popliteal arteries. 
Method; Intramuscular (IM) Gene Transfer 

Acute Limb Ischemia, Twenty-eight rabbits were used to study the impact of IM 
gene transfer on acute hindlimb ischemia. Immediately following femoral artery excision 

15 as outlined above, five different sites in three major thigh muscles were injected directly 
with plasmid DNA using a 3 ml syringe and 27-gauge needle advanced through a small 
skin incision. For each injection, the tip of the needle was inserted into the adductor (2 
sites), medial large (2 sites), and semimembranous muscles; care was taken, by directly 
visualizing each muscle during the injection, to avoid penetrating the muscle with the 

20 injectate. To the same end, the rate of injection was in each case slowed to approximately 
5 seconds so that injected solution would not leak through the epimysium. This injection 
technique was used to administer a total volume of 2.5 ml of a) a 500 ng ph VEGF165 in 
saline (n+8; b) 500 ^ig phVEGFi65 in 0.75% bupivacaine, previously shovm to enhance 
transgene uptake by striated muscle (n+10) (Danko I, Gene Therapy, 1:1 14-21 (1994); or 

25 c) 500 jxg pGSVLacZ encoding nuclear targeted p-galactosidase (n+10). After 

completing 5 injections (0.5 ml @ for each animal), the skin was then closed using 4.0 
nylon. 

Chronic Limb Ischemia. Thirty-one rabbits were used to study the effects of IM 
gene therapy for chronic hindlimb ischemia. The sole distinction between the chronic 
30 ischemia model and model of acute limb ischemia described above, is that an interval of 
10 days was permitted for post-operative recovery, including development of endogenous 
collateral vessels. Accordingly, 10 days following femoral artery excision, the rabbits 



wo 00/62798 



PCT/USOO/09765 



"33- 

were returned to the catherization laboratory. After completing baseline physiological 
measurements described below, IM gene transfer using the identical technique described 
above was performed with a) 500 \ig phVEGFies diluted in 2.5 ml of saline (n=8); b) 500 
Jig phVEGFi65 diluted in 0.75% bupivacame (n=8); c) 500 ng of pGSVLacZ diluted in 
5 2.5 ml of saline; or d) 500 iig of pGSVLacZ diluted in 2.5 ml of 0.75% bupivacaine 
(n=8). In each case, after completing all 5 injections, the skin was closed as above. 

VEGF Gene Expression in Skeletal Muscle. To evaluate expression of 
phVEGFi65 gene in skeletal muscle, sixteen additional male New Zealand white rabbits 
from both acute and chronic ischemia models (2 rabbits at each time point) were 

10 sacrificed at 3, 7, 14, and 30 days post-transfection. The presence of human VEGF 

mRNA was detected using reverse transcription-polymerase chain reaction (RT-PCR) as 
previously described (Takeshita, et al., Proc Natl Acad Sci). To ensure specificity and 
avoid amplification of endogenous rabbit VEGF, primers were selected fi-om a region 
which is not conserved among different species. Sequences of primers used were: 5'- 

15 GAGGGCAGAATCATCACGAAGT-3' (sense) (SEQ ID NO: 1): 5'- 

TCCTATGTGCTGGCCTTGGTGA-3' (antisense) (SEQ ID NO:2). RT-PCR products 
were analyzed by 2% agarose gel electrophoresis. DNA bands were visualized imder UV 
illumination after staining with ethiditun bromide. 

EXAMPLE l i Define certain morphologic, temporal, and functional aspects of 
20 therapeutic angiogenesis. 

A. Cellular proliferation contributing to the development of the nascent 
collateral circulation is augmented in ischemic limbs in response to therapeutic 
angiogenesis. 

We determined the extent to which proliferative activity of vascular cells is 
25 augmented during therapeutic angiogenesis with VEGF (Takeshita S, et al., Am J Pathol 
1995;147:1649-1660). Ten days following induction of limb ischemia by surgically 
excising femoral artery of rabbits, either VEGF (500-1,000 mg) or saline was 
administered as a bolus into the iliac artery of the ischemic limb. Cellular proliferation 
was evaluated by bromodeoxyuridine (BrDU) labeling for 24 hrs at day 0 (immediately 
30 prior to VEGF administration) and at days 3, 5, and 7 post- VEGF. EC proliferation in the 
midzone collaterals of VEGF-treated animals increased 2.8-fold at day 5 (p<0.05, vs. 
control), and returned to baseline levels by day 7. Smooth muscle cell proliferation in 



wo 00/62798 



.34. 



PCTAJSOO/09765 



midzone collaterals also increased 2.7-fold in response to VEGF (p<0.05). No significant 
increase in cellular proliferation was observed in either the stem or re-entry collaterals. 
Reduction of hemodynamic deficit in the ischemic limb measxired by lower limb blood 
pressure was docxmiented at day 7 post- VEGF (p<0.01, vs. control). These data thus 
5 support the concept that increased cellular proliferation contributes to the formation of 
collateral vessels following therapeutic angiogenesis with VEGF. 

Subsequently, we have similarly documented the time-course of cellular 
proliferative activity in a mouse model of hindlimb ischemia (CoxxfBnhal T., et al., Am J 
Pathol 1998;152:1667-1679). The femoral artery of one hindlimb was ligated and 

1 0 excised. Laser Doppler perfusion imaging (LDPI) was employed to document the 
consequent reduction in hindlimb blood flow which typically persisted for up to seven 
days. Serial in vivo examinations by LDPI disclosed that hindlimb blood flow was 
progressively augmented over the course of 14 days, ultimately reaching a plateau 
between 21 and 28 days, Morphometric analysis of capillary density performed at the 

15 same timepoints selected for in vivo analysis of blood flow by LDPI confirmed that the 
histologic sequence of neovascularization corresponded temporally to blood flow 
recovery detected in vivo. EC proliferation was documented by immimostaining for 
BrdU injected 24 hrs prior to each of these timepoints, providing further evidence that 
angiogenesis constitutes the basis for improved collateral-dependent flow in this animal 

20 model. BrdU staining was performed in the ischemic versus normal hindlimbs. 

Proliferative activity peaked at 7 days (1235 ±254 vs 8±14 BrdU-positive cells/mm^ for 
the ischemic vs normal limbs respectively (p<0.001); proliferative activity was then 
subsequently reduced at days 1 4 and 2 1 . Double immunolabeling for BrdU and CD-3 1 
demonstrated proliferating ECs in the ischemic limb. Most proliferating ECs localized to 

25 small capillaries, although EC proliferation was observed in small arteries as well. 

Capillary density and proliferative activity were also examined in mice treated with PF-4 
and sacrificed 14 days after surgery. A significant decrease in capillary density (268±1 95 
vs 1053±371 capilla^ies/mm^ p<0.01) and EC proliferation (16±29 vs 935±239 BrdU- 
positive cells/mm^, p<0.01) were found in PF-4 vs PBS-injected mice respectively. 

30 Finally, in two patients who underwent amputations followdng VEGF gene 

therapy, EC proliferative activity in the ischemic limb has been documented as well. 



wo 00/62798 PCTAJSOO/09765 

-35- 

using double immunostaining for PCNA and CD31 (Baumgartner I, et al., Circulation 
1998;97:1 1 14-1 123) (Isner JM. et al.. Lancet 1996;348:370-374). 

B. Therapeutic angiogenesis may be employed successfully to supplement 
established, native collateral circulation. 
5 Long-term experiments regarding this issue are still on-going. In the interim, 

however, we have documented by angiographic analysis supplemental collateral vascular 
development in patients with collateral vessels that had developed endogenously prior to 
gene transfer (Baumgartner I, et al., Circulation 1998;97:1 1 14-1 123) (Isner JM, et al., J 
Vase Surg 1998;28:964-975). Thus the extent of collateral development appears to be 
10 influenced less by the extent of pre-existing collaterals than by other possible 
determinants. 

C* Therapeutic angiogenesis preserves receptor-mediated endothelium- 
dependent flow in the rabbit ischemic hindlimb. 

Disturbed endothelium-dependent blood flow has been previously shown to be a 

15 feature of native collateral vessels. We therefore investigated the hypothesis that 

administration VEGF may promote recovery of disturbed endothelium-dependent blood 
flow in our rabbit model of hindlimb ischemia (Bauters C, et al., Circulation 
1995;91:2802-2809). 

Ischemia was induced by ligation of external iliac artery and excision of femoral 

20 artery in one limb of NZW rabbits (day 0). Flow velocity was measured using a Doppler 
guidewire at rest and following serotonin and acetylcholine. Blood flow (ml/min) was 
calculated assuming a circular lumen geometry. In untreated control animals with an 
ischemic limb, serotonin administered at days 10 or 40 produced a decrease in hindlimb 
blood flow (71 ±2% and 33±6% reduction from baseline, respectively); in contrast, among 

25 animals treated with a single bolus dose of VEGF administered selectively into the 
internal iliac artery at day 10 and studied at day 40, serotonin produced an increase in 
flow (1 19±8% fi-om baseline; p<0.05 vs controls). Acetylcholine induced only a moderate 
increase in flow in control animals (152±15% at day 10, 177±14% at day 40), in contrast 
to a profound increase among VEGF-treated animals studied at day 40 (254±25%; p<0.05 

30 vs controls). 

To our knowledge, these findings constitute the first demonstration of successful 
pharmacologic modulation of disturbed endothelium-dependent flow in the arterial 



wo 00/62798 PCTA;S00/09765 

-36- 

circulation subserved by collateral vessels. This physiologic benefit complements 
previously reported anatomic findings suggesting a favorable impact of angiogenic 
growth factors on collateral-dependent limb ischemia. 

EXAMPLE 2 : Investigation of certain conditional factors which may modulate the 
5 outcome of therapeutic angiogenesis. 

A. Hypoxia modulates the response to angiogenesis induced by VEGF and 

bFGF. 

To evaluate this hypothesis, we investigated whether low oxygen tension or 
cytokines known to promote neovascularization in vivo could modulate the expression of 

10 VEGF or bFGF in human vascular smooth muscle cells (SMCs) (Brogi E, et al.. 

Circulation 1994;90:649-652). SMCs were treated with platelet derived growth factor BB 
(PDGF BB) or transforming grov^ factor pi (TGF-pi) or exposed to low oxygen 
tension in serum-free medium. Northern analysis detected low basal levels of VEGF and 
bFGF mRNA in extracts of unstimulated SMCs. However, both VEGF and bFGF 

15 transcripts increased follov^dng administration of PDGF BB (10 or 20 ng/ml) or TGF-pi 
(0. 1 or 1 ng/ml). Hypoxia was a potent stimulus for VEGF gene expression, but had no 
apparent effect on bFGF steady state mRNA levels. These results documented that certain 
indirect angiogenic cytokines, such as PDGF BB or TGF-pi, may act via induction of 
bFGF and VEGF gene expression in cells resident near ECs in vivo. Hypoxia constitutes 

20 a potent stimulus for VEGF gene expression, but does not regulate bFGF under the same 
experimental conditions. 

We further investigated whether the role of human ECs might, under selected 
conditions, extend beyond that of a target to involve contingency synthesis of VEGF 
(Namiki A, et al., J Biol Chem 1995;270:31189-31 195). In both unstimulated human 

25 umbilical vein ECs (HUVECs) or human derma-derived microvascular ECs (HMECs), 
Northern analysis detected no VEGF transcripts. Phorbol-12-myristate 13-acetate (PMA, 
10*' M) treatment, however, induced VEGF mRNA expression in both HUVECs and 
HMECs, peaking at 3hrs and 6 hrs respectively, and returning to undetectable levels by 
12 hrs. In vitro exposure of HUVECs to an hypoxic environment (p02=35 nrniHg) for 

30 12, 24 and 48 hrs, and HMECs for 6, 12, 24 and 48 hrs induced VEGF mRNA in a time- 
dependent fashion. Re-exposure to normoxia (p02=l 50 mmHg) for 24 hrs after 24 hrs of 
hypoxia returned VEGF mRNA transcripts to imdetectable levels in HUVECs. Cobalt 



wo 09/62798 PCT/USOO/09765 

-37- 

chloride (Co^"*^and nickel chloride (Ni^**") treatment each induced VEGF mRNA in ECs. 
Cycloheximide treatment further augmented expression of VEGF mRNA induced by 
Co^"*", Ni^"^ and hypoxia in HUVECs. VEGF protein production in hypoxic HUVECs was 
demonstrated immunohistochemically. Conditioned media from hypoxic HUVECs 
5 caused a two-fold increase in the incorporation of tritiated thymidine. Finally, immune 
precipitates of anti-KDR probed with anti-P-tyr antibodies demonstrated evidence of 
receptor autophosphorylation in hypoxic, but not normoxic, HUVECs. These findings 
thus established the potential for an autocrine pathway that may augment and/or amplify 
the paracrine effects of VEGF in stimulating angiogenesis. 

10 To determine if ECs expressed VEGF in vivo, in situ hybridization was performed 

using a murine VEGF 1 65 cRNA probe to identify VEGF mRNA in the mouse ischemic 
hindlimb (Coufifinhal T., et si.. Am J Pathol 1998;152:1667-1679). Prior to surgery, 
scarce hybridization was detected in the ischemic limb. VEGF mRNA was also detected 
in ECs located in small capillaries or venules. In some cases, ECs of larger caliber veins 

15 also displayed positive hybridization for VEGF; VEGF expression among ECs of similar 
caliber arteries, however, was less frequent. 

Finally, we investigated the impact of hypoxia or hypoxia-dependent conditions 
on VEGF receptor expression (Brogi E, et al., J Clin Invest 1996;97:469-476). HUVECs 
and microvascular ECs (MVECs) were exposed to direct hypoxia or to medium 

20 conditioned (CM) by myoblasts maintained in hypoxia for 4 days. Control ECs were 
maintained in normoxia or normoxia-CM. Binding of *^^I-VEGF to ECs was then 
evaluated. Hypoxic treatment of ECs had no effect on '^^I-VEGF binding. However, 
treatment of ECs with hypoxia-CM produced a 3-fold increase in *^^I-VEGF binding, 
with peak at 24 h (p<0.001, ANOVA). Scatchard analysis disclosed that increased 

25 binding was due to a 13 -fold increase in KDR receptors/cell, with no change in KDR 
affinity (Kd=260+51 pM, normoxia-CM versus Kd=281j:94 pM, hypoxia-CM) and no 
change in EC number (35.6+5.9x10^ ECs/cm^ normoxia-CM versus 33.5+5.5x10^ 
ECs/cm^, hypoxia-CM). Similar results were obtained using CM from hypoxic SMCs. 
KDR upregulation was not prevented by addition to the hypoxia-CM of neutralizing 

30 antibodies against VEGF, tumor necrosis factor-a , transforming growth factor-bl or 
bFGF. Similarly, addition of VEGF or lactic acid to the normoxia-CM had no effect on 
VEGF binding. These experiments implicated a paracrine mechanism initiated by 



wo 00/62798 



-38- 



PCT/USOO/09765 



hypoxia that induces KDR receptor upregulation in ECs. Hypoxic cells, not only can 
produce VEGF, but can also modulate its effects via paracrine induction of VEGF 
receptors in ECs. 

B. Hyperchol^terolemia attenuates the response to therapeutic 
5 angiogenesis. 

We investigated the anatomic extent and functional capacity of the collateral bed 
which develops in response to limb ischemia in a well characterized animal model of 
spontaneous hypercholesterolemia, the Watanabe heritable hyperlipidemic (WHHL) 
rabbit (Van Belle E, et al., Circulation 1997;96:2667-2674\ We further characterized the 

10 impact of exogenous angiogenic cytokine administration on collateral vessel development 
and function in the same animal model. Weight-matched 6-month old male homozygous 
WHHL {n=9) and normal NZW (n=9) rabbits underwent operative resection of one 
femoral artery. Ten days later, the ischemic hindlimb was evaluated for collateral vessel 
formation, blood flow, and tissue damage. Collateral vasculature was less extensive 

15 among WHHL than NZW, as indicated by a significant reduction in angiographic score 
(0.19±0.02 vs 0.35±0.03; P < .001) and capillary density (46.4±4.1 vs 78.9±4.6/mm^, P < 
.0002). This was associated with a reduction in calf blood pressure index (9.5±3.5 vs 
32.8 ± 2.8 %, P <.0001), arterial blood flow (7.5±0.6 vs 13.6db0.7 mL/min, P <.0001), 
muscle perfusion index (40.1±3.2 vs 65.9±2.0%, P <.0001), and an increase in muscle 

20 necrosis (48.16i5.41 vs 25.90±3.83% negative 2,3,5-triphenyltetrazolium chloride 

staining, P<.004). Treatment of WHHL rabbits (n=9) with recombininant human VEGF 
produced a statistically significant improvement in all functional as well as anatomic 
indices of collateral development. Thus, collateral vessel development associated with 
hindlimb ischemia in vivo is severely attenuated in an animal model of spontaneous 

25 hypercholesterolemia, but may be nevertheless augmented by administration of 
angiogenic cytokines. 

These findings were confirmed in a murine model of hypercholesterolemia 
(Couffinhal T, et al, Circulation 1999;(In Press)), the ApoE"'* mouse, with unilateral 
hindlimb ischemia. Hindlimb blood flow and capillary density were markedly reduced in 

30 ApoE"'' mice vs C57 controls. This was associated with reduced expression of VEGF in 
the ischemic limbs of ApoE*'" mice. Cell-specific immimostaining localized VEGF 
protein expression to skeletal myocytes and infiltrating T cells in the ischemic limbs of 



wo 00/62798 PCT/USOO/09765 

-39- 

C57 mice; in contrast, T cell infiltrates in ischemic limbs of ApoE*^* mice were severely 
reduced (despite normal absolute T cell covmts in these animals). The critical 
contribution of T cells to VEGF expression and collateral vessel grov^ was reinforced 
by the finding of accelerated limb necrosis in athymic nude mice with operatively 

5 induced hindlimb ischemia. Adenoviral VEGF gene transfer to ApoE"^' mice resulted in 
marked augmentation of hindlimb blood flow and capillary density. These findings thus 
underscore the extent to which hyperlipidemia adversely affects native collateral 
development, but does not preclude augmented collateral vessel growth in response to 
exogenous cytokines. Moreover, results obtained in the ApoE"^' and athymic nude mice 

10 imply a critical role for infiltrating T cells as a source of VEGF in neovascularization of 
ischemic tissues. 

C, The response to therapeutic angiogenesis is attenuated in a diabetic 
animal model of hindlimb ischemia. 

We determined if diabetes could: 1) impair the development of new collateral 

15 vessel formation in response to tissue ischemia, and 2) inhibit cytokine-induced 
therapeutic neovascularization (Rivard A, et al,, Am J Pathol 1999;154:355-364). 
Hindlimb ischemia was created by femoral artery ligation in non-obese diabetic mice 
(NOD mice, n=20) and in control C57 mice (n=20). Hindlimb perfijsion was evaluated by 
serial laser Doppler studies after surgery. In NOD mice, measurement of the Doppler 

20 flow ratio (DFR) between the ischemic and the normal limb indicated that restoration of 
perfusion in the ischemic hindlimb was significantly impaired. At day 14 after surgery, 
DFR in the NOD mice was 0.49±0.04 vs. 0.73±0.06 for the C57 mice (p< 0.005). This 
impairment in blood flow recovery persisted throughout the duration of the study, with 
DFR values at day 35 of 0.50±0.05 vs 0.90±0.07 in the NOD and C57 mice respectively 

25 (p < 0.001). CD3 1 immunostaining confirmed the laser Doppler data by showing a 

significant reduction in capillary density in the NOD mice at 35 days after surgery (302±4 
capillaries/nmi^ vs 782i78 in C57 mice (p < 0.005). The reduction in neovascularization 
in the NOD mice was the result of a lower level of VEGF in the ischemic tissues, as 
assessed by Northern blot, Westem blot and immunohistochemistry. The central role of 

30 VEGF was confirmed by showing that normal levels of neovascularization (compared to 
C57) could be achieved in NOD mice that had been supplemented for this growth factor 
via IM injection of an adenoviral vector encoding for VEGF. We concluded that: 1) 



wo 00/62798 PCTA:S00/09765 

-40- 

diabetes impairs endogenous neovascularization of ischemic tissues; 2) the impairment in 
new blood vessel formation results from reduced expression of VEGF; and 3) cytokine 
supplementation achieved by IM adeno-VEGF gene transfer restores neovascularization 
in a mouse model of diabetes. 

5 EXAMPLE 3 ; The impact of growth factor selection, mode of delivery, and use of 
adjunctive therapies in optimizing the anatomic and physiologic outcomes of 
therapeutic angiogenesis. 

A. The character and magnitude of cellular proliferation observed in 
response to VEGF may be modified by co-administration of a second angiogenic 

10 growth factor. 

To test this hypothesis, we evaluated the extent of neovascularization which was 
achieved in vivo following intra-arterial administration of VEGF (500 |ig) alone, bFGF 
(10 ^g) alone, and VEGF(500 ng)+bFGF(10^g) all as recombinant protein, to the 
intemal iliac artery of the rabbit ischemic hindlimb (Asahara T, et al., Circulation 

15 1995;92:II-365-II-37l\ Augmentation of calf blood pressure ratio as well as papaverine- 
induced maximum flow reserve was significantly (p<0.05) greater in the VEGF+bFGF 
group than the VEGF, bFGF, or saline control groups. The extent of neointimal 
thickening in the intemal iliac artery at day 30 was not significantly different among the 
foiu- experimental groups. This study thus demonstrated that co -administration of VEGF 

20 and bFGF produces a greater (and parenthetically more rapid) improvement in vascularity 
than either VEGF or bFGF alone. 

We have also demonstrated that the pleiotropic effects of certain grov^ factors 
may potentiate angiogenesis due to a combination of direct effects on EC proliferation 
and migration, and indirect effects that result in the generation of other potent EC 

25 mitogens from non-EC populations (Van Belle E, et al.. Circulation 1998;97:381-390). 
In the case of hepatocyte growth factor (HGF), the synergistic effect, which results from 
simultaneous administration of VEGF in vitro, is reproduced in vivo by HGF-induced 
upregulation of VEGF in vascular SMCs. HGF is a pleiotropic growth factor, which 
stimulates proliferation and migration of ECs via the c-Met receptor, present on ECs as 

30 well as other cell types, including SMCs. We studied the effects of recombinant human 
(rh) HGF in vitro, and in vivo in our rabbit model of hindlimb ischemia. We further 
compared these effects with those of recombinant human VEGF (rhVEGFiss). 



wo 00/62798 



PCT/USOe/09765 



-41- 

In vitro, rhHGF and rhVEGFies exhibited similar effects on proliferation and 
migration of ECs. When both cytokines were administered together, the result was an 
additive effect on EC proliferation, and a synergistic effect on EC migration. Application 
of rhHGF to cultures of human SMCs resulted in the induction of VEGF mRNA and 

5 protein. In vivo, administration of rhHGF (500 [ig X 3) was associated with significant 
improvements in collateral formation (p<.001) and regional blood flow (p<.0005), and 
with a significant reduction in muscle atrophy (p<.0001). These effects were significantly 
more pronoxmced than those of rhVEGFi65 administered according to the same protocol 
(p<.05). Neither remote angiogenesis nor other pathologic sequellae were observed with 

10 either rhHGF or rhVEGFi65. Thus, the finding of a potentiated angiogenic effect of 
rhHGF via induction of VEGF constitutes what is in essence paracrine amplification of 
angiogenesis 

Finally, we tested the hypothesis that gene transfer of plasmid DNA encoding 
angiopoietinl (Angl) and Ang2 could modulate collateral vessel development in a rabbit 

15 model of hindlimb ischemia (Shyu K-G, et al.. Circulation 1998;98:2081-2087). Angl, 
but not Ang2, gene transfer produced anatomic and physiologic evidence of enhanced 
collateral vessel formation because Angl is known not to have any effect on EC 
proliferation, it is possible that a synergistic effect between exogenous Angl and 
endogenous VEGF expression accounts for the finding of enhanced collateral 

20 development in response to Ang 1 . 

B. The magnitude of angiogenesis developing in response to administration 
of exogenous growth factors may be augmented by administration of heparin. 

Ten days after excision of the femoral artery in one limb of NZW rabbits, heparin 
(800 lU, n=13), VEGF (1 mg, n=3; 5 mg, n=5), heparin (800 lU) + VEGF (1 mg, n=5; 5 

25 mg, n=7), or saline (n=8) was injected as a single bolus in a marginal ear vein (Banters C 
et al., J Vase Surg 1 995 ;2 1 :3 14-325). Collateral vessel formation and limb perfiision were 
assessed 10 and 30 days after treatment. Animals in both VEGF-treated groups had a 
significantly higher (p <0.01) increase in calf blood pressure ratio at day 10 (control = 
0.44±0.02; heparin = 0.47±0.02; VEGF = 0.60±0.01; [heparin + VEGF] = 0.61±0.02) and 

30 day 30 (control = 0.49±0.05; heparin = 0.48±0.02; VEGF = 0.70db0.03; [heparin + VEGF] 
= 0.73±0.03). Both VEGF-treated groups had a significantly higher (p <0.05) 
angiographic score at day 30 (control = 0.28±0.01; heparin = 0.28±0.01; VEGF = 



wo 00/62798 PCT/US00/0976S 

-42- 

0.37±0.01 ; [heparin + VEGF] = 0.38±0.02). Maximum flow reserve at day 30 in the 
ischemic limb was higher (p <0.05) in VEGF-treated rabbits (control = 1 .87±0.07; 
heparin = 1.92±0.08; VEGF = 2.42±0.16; [heparin + VEGF] = 2.33±0.12). Capillary 
density was higher (p <0.01) in the ischemic muscles of VEGF-treated rabbits (control = 
156±10/nim^ heparin = 178±8/mm^ VEGF = 230±10/mm^ [heparin + VEGF] = 
233±8/nmi^). This series of in vivo experiments demonstrated that intravenous 
administration of VEGF, vwth or without heparin, results in both anatomic and 
physiologic evidence of enhanced collateral vessel formation in the rabbit ischemic 
hindlimb. 

C, Coincident activation of plasminogen facilitates therapeutic angiogenesis. 

The requirement that ECs must remove certain constraining physical influences, 
including the attachment to their underlying basement membrane and the more peripheral 
barrier posed by their extracellular matrix, to facilitate cell movement for development of 
a neovascular sprout, represents for angiogenesis a fundamental tenet (Vernon RB, et al.. 
Am J Pathol 1995;147:873-883). Houck et al (Houck KA, et al., J Biol Chem 
1992;267:26031-26037) previously reported that cleavage by plasmin of VEGF189 at its - 
COOH terminus generates a 34 kD proteolytic fi-agment (cl-VEGFigp) which is mitogenic 
for ECs and active as a permeability agent. Park et al (Park JE, et al., Mol Biol Cell 
1993;4:1317-1326) have reported that longer forms of VEGF are stably incorporated into 
the extracellular matrix, but can become available in diffusible form when the matrix is 
degraded by plasmin. We are currenUy investigating the possibility that coordinated 
extracellular matrix degradation - achieved by generating plasmin via administration of 
recombinant t-PA or co-transfection of plasmid cDNA encoding t-PA - may facilitate 
therapeutic angiogenesis. 

D. The magnitude of the angiogenic response does not vary as a function of 
the distance from the ischemic site at which VEGF is administered. 

The series of in vivo experiments described above (Example 3,B) established that 
intravenous administration of VEGF, with or without heparin, results in both anatomic 
and physiologic evidence of enhanced collateral vessel formation in the rabbit ischemic 
hindlimb (Banters C, et al., J Vase Surg 1995;21 :3 14-325). A similar series of 
experiments demonstrated that IM administration of VEGF recombinant protein 
improved hindlimb perfusion to a similar extent as was seen vwth intra-arterial (and 



wo 00/62798 



^3- 



PCT/USOO/09765 



intravenous) delivery of the protein (Takeshita S, et al.. Circulation 1994;90:II-228-II- 
234). 

E. The magnitude of angiogenesis observed in response to VEGF varies as 
a function of the isoform of VEGF employed. 

5 Plasmid DNA encoding each of the three principal human VEGF 1 (VEGF A) 

isoforms (phVEGFai, phVEGFi65, or phVEGFigg) was apphed to the hydrogel polymer 
coating of an angioplasty balloon, and delivered percutaneously to one iliac artery of 
rabbits with operatively induced hindlimb ischemia (Takeshita S, et al., Lab Invest 
1996;75;487-502). Compared to control animals transfected with LacZ, site-specific 

10 transfection of phVEGF resulted in augmented collateral vessel development documented 
by serial angiography, improvement in calf blood pressure ratio (ischemic/normal limb), 
resting and maximum blood flow, and capillary/myocyte ratio. Similar results were 
obtained with phVEGF^i, phVEGFies, and phVEGFi89, suggesting that these isoforms 
are biologically equivalent with respect to in vivo angiogenesis. The fact that viral or 

15 other adjunctive vectors were not required further suggests that secreted gene products 
may have potential therapeutic utility even when the number of successfully transfected 
cells remains low. Arterial gene transfer of naked DNA encoding for a secreted 
angiogenic cytokine thus represents a potential alternative to recombinant protein 
administration for stimulating collateral vessel development. 

20 F. Therapeutic angiogenesis may be effectively performed using direct 

intramuscular (IM) administration of the gene encoding VEGF. 

Striated muscle had been shown to be capable of taking up and expressing foreign 
genes transferred in the form of naked plasmid DNA, though typically with a low level of 
gene expression. We had shown, however, that in the case of genes which encode 

25 secreted proteins, low transfection efficiency did not preclude bioactivity of the secreted 
gene product (Takeshita S, et al.. Lab Invest 1994;71:387-391) (Losordo DW, et al., 
Circulation 1994;89:785-792). Accordingly, we investigated the hypothesis that IM gene 
therapy with naked plasmid DNA encoding VEGF could augment collateral development 
and tissue perfusion in the rabbit ischemic hindlimb (Tsurumi Y, et al.. Circulation 

30 1996;94:3281-3290). 

Ten days after ischemia was induced in one rabbit hindlimb, 500 \ig of 
phVEGFi65, or the reporter gene LacZy were injected IM into the ischemic hindlimb 



wo 00/62798 



-44- 



PCTAJSOO/0976S 



muscles. Thirty days later, angiographically recognizable collateral vessels and 
histologically identifiable capillaries were increased in VEGF-transfectants compared to 
controls. This augmented vascularity improved perfusion to the ischemic limb, 
documented by a superior calf blood pressure ratio for phVEGFiss (0.85i0.05) vs. 

5 controls (0.64±0.05, p<.01); improved blood flow in the ischemic limb (measured using 
an intra-arterial Doppler wire) at rest (phVEGFi65=21.3±3.9, control=14.6±1.6 ml/min, 
p<.01) and following a vasodilator (phVEGFi65=54.2±12.0, control=37.3±8.9, p<.01); 
and increased microspheres in the adductor (phVEGFi65=4.3i:1.6, control=2.9±1.2 
ml/min/lOOg tissue, p<.05), and gastrocnemius (phVEGFi65=3.9dbl.O, control=2.8±1.4 

10 ml/min/ lOOg tissue, p<.05) muscles of the ischemic limb. These experiments established 
that ischemic skeletal muscle constituted a promising target for gene therapy with naked 
plasmid DNA. IM transfection of genes encoding angiogenic cytokines, particularly 
those which are naturally secreted by intact cells, thus appeared to represent an alternative 
treatment strategy for patients in whom extensive vascular disease prohibited access to 

15 the lower extremity vasculature otherwise required for intra-arterial catheter-based gene 
transfer. 

Based on this animal data, we initiated a phase I clinical trial to (1) document the 
safety and feasibility of IM gene transfer using naked plasmid DNA encoding VEGF, and 
(2) analyze potential therapeutic benefits in patients with CLI (Baumgartner I, et al., 

20 Circulation 1998;97:1 1 14-1 123) (Isner JM, et al., J Vase Surg 1998;28:964-975). 

Gene transfer was performed in 1 0 limbs of 9 patients with non-healing ischemic 
ulcers (n=7/10) and/or rest pain (n=10/10) due to peripheral arterial disease. A total dose 
of 4000 ^ig of phVEGFi65 was injected directly into the muscles of the ischemic limb. 
Gene expression was documented by a transient increase in serum levels of VEGF 

25 monitored by ELISA assay. The ankle-brachial index improved significantly (0.33 ± 
0.05 to 0.48 ± 0.03, p=0,02), new collateral blood vessels were directly visualized by 
contrast angiography in 7 limbs, and magnetic resonance angiography showed qualitative 
evidence of improved distal flow in 8 limbs. Ischemic ulcers healed or markedly 
improved m 4/7 limbs, including successful limb salvage in 3 patients recommended for 

30 below-knee amputation. Tissue specimens obtained from an amputee 10 wks after gene 
therapy showed foci of proliferating ECs by immunohistochemistry. PGR and Southern 
blot analyses indicated persistence of small amounts of plasmid DNA. Complications 



wo 00/62798 



PCTAJS00/0976S 



-45- 

were limited to transient lower extremity edema in 6 patients, consistent with VEGF- 
enhancement of vascular permeability. 

These findings thus demonstrated that IM injection of naked plasmid DNA may 
achieve constitutive overexpression of VEGF sufficient to induce therapeutic 
5 angiogenesis in selected patients with CLI. 

Most recently, we adapted the use of IM phVEGFies gene transfer to investigate 
gene therapy for therapeutic angiogenesis in patients with myocardial ischemia (Losordo 
DW, et al.. Circulation 1998;98:2800-2804). A phase 1 clinical study was initiated to 
determine the safety and bioactivity of direct intramyocardial gene transfer of VEGF as 
10 sole therapy for patients with symptomatic myocardial ischemia. 

VEGF gene transfer (GTx) was performed in 5 patients (all male, ages 53-71) 
with angina due to angiographically documented coronary artery disease (CAD) who had 
failed conventional therapy (drugs, angioplasty surgery). Naked plasmid DNA encoding 
VEGF (phVEGFi65) was injected directly into the ischemic myocardiimi via a "mini" left 
15 anterior thoracotomy. Injections caused no changes in heart rate (pre-GTx=75±15/min vs 
post-GTx=80±16/min, p=NS), systolic BP (1 14±7 mmHg vs 118±7mm Hg, p=NS), or 
diastolic BP (57±2 mmHg vs 59±2 nrmiHg, p=NS). Ventricular arrhythmias were limited 
to single unifocal premature beats at the moment of injection. Serial ECGs showed no 
evidence of new myocardial infarction in any pt. Intraoperative blood loss was 0-50cc 
20 and total chest tube drainage was 1 10-395cc. Cardiac output fell transiently post-op but 
increased within 24 hrs (pre-anesthesia = 4.8±0.4 vs post-anesthesia = 4.1±0.3 vs 24 hrs 
post-operatively = 6.3±0.8, p=0.02). Time to extubation following closure was 18.4±1 .4 
min and avg post-op hospital stay was 3.8 days. All patients had significant reduction in 
angina (NTG use = 53.9+10.0/wk pre-GTx vs 9.8±6.9/wk post-GTx, p<0.03). Post- 
25 operative left ventricular ejection fraction (LVEF) was either unchanged (n=3) or 

improved (n=2, mean increase in LVEF = 5%). Objective evidence of reduced ischemia 
was documented using dobutamine SPECT-sestamibi imaging in all patients. Coronary 
angiography showed improved Rentrop score in 5/5 patients. 

This initial experience with naked gene transfer as sole therapy for myocardial 
30 ischemia suggested that direct intramyocardial injection of naked plasmid DNA via a 
minimally invasive chest wall incision is safe and may lead to reduced symptoms and 
improved myocardial perfusion in selected patients with chronic myocardial ischemia. As 



wo 00/62798 



^6- 



PCT/USOO/09765 



of February, 1999, a total of 23 patients have been treated in this fashion with similar 
results (Symes JF, et al., Ann Thorac Surg 1999). 

In summary, the above-described experiments extended the notion of therapeutic 
angiogenesis from in vitro studies and animal models to patients with lower extremity 
5 and myocardial ischemia. 

EXAMPLE 4 : Peripheral Neuropathy 

!• Neurological findings in patients undergoing phVEGFi^sgene transfer. 

Methods and results: We prospectively evaluated neurological and 
neurophysiological findings in patients undergoing phVEGFies gene transfer for critical 

10 limb ischemia (CLI). All patients were evaluated by two neurologists, one performing 
clinical assessment, and one performing electrophysiologic testing. Both were blinded to 
each other's results, and both were blinded to the results of patients' vascular 
examinations. Furthermore, at follow-up examinations, both were blinded to results of 
previous examinations. Quantitative sensory testing was performed using CASE IV 

15 (Computer Aided Sensory Evaluator, a quantitative sensory testing device for thermal 
pain and vibration thresholds (Dyck PJ, et al., Diabetes Care 1987;10:432-440)). Tibial 
motor, peroneal motor, and sural sensory electrophysiologic studies were performed 
using standard techniques. Both lower extremities were studied, except in patients in 
whom the contralateral, non-treated limb was not available due to previous amputation. A 

20 total of 24 limbs have thus far been analyzed before and 3 mo after gene transfer; 19 of 
these have been followed to 6 months. 

These findings suggest that therapeutic angiogenesis have a favorable impact on 
established ischemic peripheral neiiropathy. With regard to symptoms, for example, as 
early as 3 mo. Symptom Score, encompassing 5 neuropathy-related symptoms, decreased 

25 (i.e. improved) from (m=bSEM) 3.3±0.5 to 1.7±0.4 (p<0.01). Sensory Disability Score 
decreased from 9.5±1 .3 to 7.4±1.2 (p<0.01). By 6 mo. Symptom Score decreased to 
1 .1±0.4 (p<0.01 vs baseline of 3.4). Sensory Disability Score likewise decreased to 
6.3±1.4 (p=0.01 vs baseline 9.5±1.3), along with a reduction in Total Disability Score 
(12.7±2.1 to 9.2±1.7, p=0.01). Vibration threshold decreased (i.e. improved) fi:om 

30 21 .0±0.9 to 19.8il .0, p=0,04. Moreover, by 6 months, improvement in objective indices 
of nerve function became manifest. Peroneal motor nerve amplitude increased from 



wo 00/62798 



-47- 



PCT/USOO/09765 



2. liO.6 to 2.8i:0.6 (p=0.03). In no case did clinical examination or electrodiagnostic 
studies show statistically significant improvement for measurements 
recorded from the contralateral, non-treated lower extremity. The percent change in 
peroneal motor amplitude in the treated vs untreated leg for each of the 19 patients 
5 followed for 6 months (one patient, who began with an amplitude of 0 and increased to 
0.2 is not shown); illustrates that increases were observed in three insulin-requiring 
diabetics (patients 1,5, and 11). Also included was one patient in whom we documented 
the appearance of a previously absent potential. For the group as a whole, the percent 
increase in peroneal amplitude (50.3±21.0) exceeded the change observed in the non- 

10 treated leg (-10.8^7.9) to a statistically significant degree (p=0.02). Similarly, the simi of 
motor amplitude increased 16.5±8.7% vs the non-treated leg (-8.8±6.5), (p=0.04). 

Conclusions: This prospective study - the first to our knowledge to investigate 
the impact of therapeutic angiogenesis on peripheral nerve function in patients with limb 
ischemia - showed evidence of improvement in peripheral nerve function in patients 

1 5 undergoing phVEGF i es gene transfer. 

2. Adaptation of the rabbit ischemic hindlimb model for the investigation of 
ischemic peripheral neuropathy. 

Methods and results: The rabbit model of hindlimb ischemia has been 
extensively characterized in our laboratory and frequently utilized to investigate strategies 

20 of therapeutic angiogenesis. We therefore xmdertook a series of preliminary experiments 
to determine the extent of peripheral neuropathy which might accompany the 
development of vascular insufficiency in this model. 

Determination of normal electrophysiologic parameters. All protocols were 
approved by the Institutional Animal Care and Use Committee at St. Elizabeth's Medical 

25 Center. Male NZW rabbits 5-6 yrs of age (mean weight = 5 kg) were used for all 

experiments. Pilot experiments performed in our laboratory showed that when hindlimb 
ischemia is created in young (6-8 mo) NZW rabbits, development of neuropathy is 
transient and of unpredictable magnitude. We therefore performed pilot experiments in 
old (5 yrs) rabbits, in which angiogenesis was shown to be retarded (Rivard A, et al., 

30 Circulation 1 999;99:3 11-1 20) and documented persistent and profound neurophysiologic 
abnormalities. Before recordings from ischemic limbs could be made, it was necessary to 
document the electrophysiologic responses recorded from the rabbits' non-ischemic 



wo 00/62798 PCT/US00/0976S 

-48- 

hindlimb. Motor and sensory nerve potentials were recorded from both limbs of 8 rabbits 
with intact femoral arteries. Data were considered as mean ± standard error of the 
(m±SEM).Compound muscle action potentials (CMAPs) were 16.0±1.2 mVin the left 
limb and 16.4±0.9 mV in the right limb (p=ns). Sensory nerve action potentials (SNAPs) 
5 were 7.7±0.8 ^V in the left limb vs 8. 1±1 . 1 jiV in the right limb (p=ns). 

(Neurophysiologic recordings in the non-ischemic limbs of old rabbits were not different 
from those recorded in non-ischemic limbs of yoimg rabbits). 

Induction of ischemia in the rabbit hindlimb. Unilateral hindlimb vascular 
insufficiency was operatively created in 10 rabbits using previously published techniques 

1 0 (Takeshita S, et al., J Clin Invest 1 994;93 :662-670). 

Verification of ischemic peripheral neuropathy. After surgical induction of 
unilateral hindlimb ischemia, rabbits were allowed to recover for 5 days. 
Electrophysiologic recording of peripheral nerve function in the ischemic and non- 
ischemic limbs was then performed xmder general anesthesia on a weekly basis for 3 

15 months CMAPs and SNAPs were recorded. Motor nerve conduction velocities (MCV) 
and sensory nerve conduction velocities (SCV) were calculated. Concomitantly, the calf 
blood pressure ratio, defined as the ratio of systolic pressure of the ischemic limb to that 
of the normal limb, was determined for each rabbit using standard techniques. At the 
defined end point, 12 wks after induction of ischemia, rabbits also underwent selective 

20 angiography of the internal iliac artery as previously described (Takeshita S, et ah, 7 C/m 
/rtve5M994;93:662-670). 

The development of hindlimb ischemia had a profound effect on peripheral nerve 
function. Data were considered to represent function in right (ischemic) limb as a 
percentage of that recorded in left (non-ischemic) limb. Motor nerve amplitudes, 

25 indicated by CMAP, dropped to zero post-operatively before they became detectable 

again 4 wks later; at 8 wks post-operatively, CMAPs were still only 10 to 15% of normal. 
From wk 8 on, CMAPs increased in a nearly linear fashion, improving to 50-60% of 
normal by wk 10, vwth no further changes through wk 12 (56.2±7.3%). MCV behaved 
similarly: ischemic limbs displayed non-determinable conduction velocities up to wk 3. 

30 From wk 4 on, MCV increased in a nearly linear pattern, peaking at 87.4±3.6% of normal 
by wk 12. 



wo 00/62798 PCT/USOO/09765 

-49- 

In contrast to motor nerve parameters, SNAPs did not drop to zero level following 
induction of ischemia, but did remain depressed at 1 7 - 22% of normal between wks 1 to 
4 following induction of ischemia. From then on, SNAPs increased to 51.7±5.1% of 
normal at wk 12. In contrast to MCV and CMAP, which were undetectable after the 

5 induction of ischemia, SCVs were initially reduced to approximately 20%, before they 
started to rise at wk 3 and by wk 12 were 98.9±0. 1% of normal. Ischemia reduced calf 
blood pressure ratio to 22.9±5.4% at wk 1 . An almost linear increase in calf blood 
pressure ratio peaked at 79.0±9.0% at wk 12. Finally, morphometric angiographic 
analysis, performed at wk 12, revealed that the angiographic score (quantitative analysis 

1 0 of vascular density based on contrast angiograms for ischemic limbs was 44.8±4.5% of 
non-ischemic limbs, consistent with endogenous neovascularization described previously 
for this model (Takeshita S, et al., J Clin Invest 1994;93:662-670). Pathologic findings 
included focal loss of axons and disrupted myelin sheaths. 

Conclusion: These findings indicate that hindlimb ischemia causes a severe 

15 peripheral neuropathy which affects both motor and sensory nerve functions. The rabbit 
model of hindlimb ischemia thus represents a suitable tool for the investigation of 
ischemic peripheral neuropathy. 



20 

3. Therapeutic angiogenesis attenuates the development of ischemic 
peripheral neuropathy. 

Methods and results. Previous work from our laboratory has demonstrated that 
IM phVEGFi65 gene transfer performed at the time of siugery to create unilateral 

25 hindlimb ischemia results in accelerated revascularization of the ischemic hindlimb, in 
comparison to control animals injected with a reporter gene (Tsurumi Y, et al., 
Circulation 1996;94;328 1-3290). Accordingly, we performed a pilot study involving 10 
rabbits, each of which received 5 injections of phVEGFi65 (100 fig @) into the adductor 
(2 sites), medial large (2 sites), and semimembranosus (one site) muscles at the time the 

30 animals imderwent unilateral excision of the femoral artery to create hindlimb ischemia. 
Following surgery and gene transfer, rabbits underwent electrophysiologic and vascular 
examinations on a weekly basis. Immediately prior to sacrifice at 12 wks, selective 



wo 00/62798 PCTAJSOO/05765 

-50- 

angiography was performed. Postmortem examination included analysis of 
capiliary/myocyte ratios and morphometric examination of nerve tissue sections. 

One week post-surgery, CMAPs recorded in the ischemic limb were reduced to 
36.8±9.1% of that recorded from the left limb. At wk two, however, CMAPs had 
5 improved to 49.9±0.5% of normal, and by 3 wks improved further to 82.0i:3.4% of 
normal. CMAP ratios for wks 8 to 12 displayed a plateau-like pattern with values 
ranging from 83.9dbl .3% at wk 8 to 90,5±4.3% at wk 12. In comparison, CMAPs for the 
controls were 10.9±2.9% of normal at wk 8, and 56.2±7.3% at wk 12. From wk 8 to wk 
12, CMAPs were significantly higher at each timepoint in day 0-treated rabbits. 

10 Measurements of MCV demonstrated a moderate drop in conduction velocity for 

a brief period of two wks after the induction of ischemia (83. 5^1 1 .0% and 89.9±7.1% at 
wks 2 and 3, respectively). MCVs at these 2 timepoints were significantly different from 
values in the non-ischemic limb. Thereafter, however, MCV recovered to normal values, 
from wk 4 through wk 12. 

15 Examination of sensory nerve ftmction disclosed a reduction in SNAPs from 

100% at baseline to 70.2±1 1.3% within the first wk after surgery. This parameter 
continued to be significantly lower than normal up to wk 4. Neurophysiological 
assessment at wk 5, however, disclosed improvement in SNAPs to 99.6dbl6.6%, hence no 
longer significantly different from normal baseline values. No fiirther significant change 

20 in SNAPs was observed up to wk 12 when the animals were sacrificed. 

SCVs were also recorded in this study group and were foimd to be only slightly 
depressed within the first wks after the onset of ischemia. The lowest SVC was 
90.6±7.3% at wk 3. All other timepoints were even closer to normal or absolutely normal, 
and none of the values taken differed significantly from normal vdues. 

25 At each timepoint employed for neurophysiological examinations, blood pressure 

ratios for the rabbits' hindlimbs were recorded as well. The results with the motor nerve 
parameters show that post-operatively there was an expected drop in the blood pressure 
ratio to 38.0±9.1% of normal. Three wks post-operatively, the calf blood pressure ratio 
had risen again to 82.9±4.7%. Further follow-up examinations showed a progressive rise 

30 in blood pressure ratio to 97.2±1 . 1% at wk 12 before sacrifice. Pathologic observations 
indicated improved preservation of axons and myelin sheaths, compared to imtreated 
animals. 



wo 00/62798 PCTAJSCO/09765 

-51- 

Conclusion: Day-0 IM gene transfer of phVEGFi6s prevented CMAPs as well as 
SNAPS from dropping to unrecordably low levels, and led to significantly faster recovery 
of CMAPs and SNAPs. An even more obvious impact of treatment was seen on MCV 
and SCV, both of which were only mildly reduced within the first 3 wks after ischemia. 
5 Improvement in calf blood pressure ratio paralleled these neurophysiologic findings. 

4. Therapeutic angiogenesis promotes recovery of established ischemic 
peripheral neuropathy. 

Methods and results: To investigate whether established ischemic neuropathy can 
be reversed by therapeutic angiogenesis, 10 rabbits underwent excision of the femoral 
10 artery followed by a 10 day interval to permit development of severe hindlimb ischemia 
and hence ischemic neuropathy. At day 10, rabbits underwent phVEGFies gene transfer 
for therapeutic angiogenesis as outlined above. Neuroelectrophysiologic and blood 
pressure measurements were again performed on a weekly basis according to the protocol 
described above. 

15 Within the 10-day period between induction of ischemia and treatment, CMAPs 

dropped to undetectable levels. At week 4 (2.5 wks post-treatment), amplitudes were 
16.8±1.6% of baseline. Subsequent follow-up examinations documented improvement in 
CMAPs, increasing to 44. ld:4.7% at wk 6, and 69. 1±2.2% at the endpoint (wk 12 after 
the induction of ischemia, i.e. 10.5 wks post-gene transfer). 

20 MCVs were detectable neither at first examination after excision of the femoral 

artery, nor briefly after gene therapy. However, by wk 3 (1 .5 wks after gene transfer), 
MCV increased to 35.0±14.1% of normal, and one wk later to 74.8±3.2% of normal. By 
wk 12, MCV in the ischemic limb was restored to 86.5±3.9% of normal. 

SNAPs decreased to 38.3 ± 15.2% of normal following induction of ischemia. 

25 The first significant change in SNAPs thereafter was detectable 2.5 wks after gene 
therapy (wk 4), when the SNAP ratio improved to 64.9±8.4%. By wk 6, SNAPs had 
reached normal values again (103.3±10.5%), and at wk 12 were 96.9±4.9%. 

SCVs, in contrast to the MCVs, were less severely affected by induction of 
ischemia: no significant alteration (in comparison to the contralateral non-ischemic 

30 contralateral limb) could be detected. 

Blood pressure measurements in this study group disclosed the anticipated 
reduction in limb perftision post-surgery (blood pressure ratio in the ischemic to normal 



wo 00/62798 PCTA;S00/09765 

-52- 

limb = 21.8±5.4%). Following gene therapy, blood pressure ratio showed an almost 
linear increase, peaking at 95.2 ±0.3% by wk 12. 

Conclusion: For SNAPs, the day O-treated group showed the most favorable 
outcome in terms of minimally impaired nerve function and quick recovery. In the day 
5 1 O-treated group, however, SNAPs were also restored earlier, reaching normal function 
again by 4.5 wks after gene therapy. SNAPS in untreated animals remain reduced 
(51.7±5.1% of normal values). CMAP recovery of day O-treated animals was fastest and 
most complete, whereas onset of recovery in xmtreated rabbits was delayed to 6.5 wks 
after surgery. CMAPs in the day 10-treatment group recovered faster than those in 

10 untreated rabbits. These findings suggest a favorable effect of therapeutic angiogenesis on 
the development of ischemic peripheral neuropathy. 

5. Expression of VEGF and VEGF receptor by Schwann cells in vitro 
Methods and results: NZW rabbits were sacrificed under aseptic conditions using 
an intracardiac injection of pentobarbital sodium, following which their sciatic nerves 

15 were dissected free, harvested, and processed as previously described (Morrissey TK, et 
al., JNeurosci 1991 ;1 1 :2433-2442). Briefly, the nerves were placed into Liebovitz's L-15 
medium (GIBCO) supplemented with 50U/ml penicillin and 0.05 mg/ml streptomycin (L- 
15a). The epineurium, connective tissue, and blood vessels were stripped off with fine 
forceps. The nerves were placed in fresh L-15a and minced with scissors or scalpel blades 

20 into roughly 1x1 mm^ explants. Minced nerves were placed in a 35-mm dish containing 
1.25 U/ml dispase (Boehringer Mannheim Biochemicals), 0.05% collagenase 
(Worthington Biochemicals), 20% fetal bovine serum (FBS), and 25 mM HEPES in 
Dulbecco's Modified Eagle's Mediiun (DMEM,GIBCO). The tissue in this solution was 
triturated in a Pasteur pipette approximately 10 times and was then incubated overnight at 

25 37**C in a humidified atmosphere of air with 5% CO2. The following day, the explants 
were dissociated by gentle trituration through a flame-narrowed borosilicate pipette (0.5 - 
1-mm bore), washed 2-3 times in DMEM/10% FCS, and seeded into poly-D-lysine- 
coated (Sigma) tissue culture flasks (FALCON). To amplify the Schwann cell cultures, 
the cells were grown on poly-D-lysine-coated tissue culture plastic in DMEM 

30 supplemented with 10% FBS, 2 ^M forskolin (Sigma) and 10 ^g/ml bovine pituitary 
extract (GIBCO). 



wo 00/62798 PCT/US00/0976S 

.53- 

Schwann cells were identified in cultures on the basis of cell soma and nuclear 
morphology lising phase microscopy. Cells with long hi- or tripolar processes (that were 
approximately 5 -7 times the width of the cell body) and oval nuclei were counted as 
Schwann ceils. In fixed cultures, we performed immunocytochemical labeling for SI 00 
5 protein. Cells were fixed for 10 min in 4% paraformaldehyde followed by 

permeabilization in 4% paraformaldehyde with 0.02% Triton X-100. After blocking with 
L- 15/10% heat-inactivated horse serum, the cells were incubated with a mouse 
monoclonal anti-SlOO (b-subunit) antibody (1:1000; Sigma) overnight at 4°C. The 
following day, the cells were fiirther processed by incubation with a biotinylated 

10 secondary horse anti-mouse antibody for 30 min at room temperature. Slides were then 
labeled with an Ultra Streptavidin Enzyme Complex (Signet Laboratories) according to 
the manufacturer's directions. 

Schwann cell expression of VEGF and Flt-1 (VEGF receptor 1, or VEGFR-1) 
protein expression was assessed by Western blotting. Cells were lysed by addition of 1 

1 5 ml RIPA buffer (1% NP-40, 0.5% sodium deoxycholic acid, 0. 1 % SDS in PBS, pH 7.4; 1 
^iM leupeptin; 5 \iM aprotinin; 1 mM PMSA; and 1 jiM pepstatin, all Sigma) per 100- 
mm plate. Protein extracts (100 ng) were separated on a 10% SDS-PAGE and transferred 
to a 0.2-fim PVDF membrane (Bio Rad). The membranes were blocked in 10% nonfat 
dry milk/0.2% Tween-20 in PBS, pH 7.4, then immunoblotted with a mouse monoclonal 

20 anti-human VEGF antibody (1 :250) or a mouse monoclonal anti-human Flt-1 antibody 
(1 :500) (both antibodies firom Sigma) overnight at 4°C. Blots were washed with 0.2% 
Tween-20 in PBS and incubated with horseradish peroxidase-linked goat anti-mouse 
antibody (1:10000; Sigma) for 45 min. Imunoreactive bands were visualized with ECL 
reagent (Amersham). Very recent data from our laboratory has identified mRNA for both 

25 VEGFR2 (KDR) and neuropilin-1 in cultured Schwann cells as well. 

We also assessed Schwann cell VEGF expression by ELISA testing upon the 
culture mediimi of equal numbers of cells in 24-well tissue culture plates. After the cells 
had grown to confluence, they were washed with DPBS and then further kept in DMEM 
containing various concentrations of cytokine- and growth factor-fi'ee defined FBS. After 

30 24 hrs of incubation, equal voliunes of supematants from each test condition were 

removed, and the samples were cleared firom cell debris by centrifiigation (12000 rpm for 
5 min). VEGF protein was determined with an immunoassay according to the 



wo 00/62798 PCT/USOO/09765 

-54- 

manufacturer's instructions (R&D Systems). Results were compared with a standard 
curve of human VEGF with a lower detection limit of 5 pg/ml. Samples were checked by 
serial dilution and were performed in duplicate. The results are shown in Example Table 
1. 

5 Example Table 1. ELISA measurement of VEGF protein (n=2) for each condition 





Defined FBS content in media 




0.5% 1% 5% 10% 




• VEGFi65 (pg/nal) 


Control 
medium 


12.6 


9.2 


1.1 


63.4 


Cell culture 
supernatant 


851.5 


759.8 


1339.4 


1935.5 



Conclusion: Schwann cells express VEGF protein as well as VEGFR-1 (Flt-1) in 

vitro. 

6. Schwann cell migration in response to VEGF 

10 Methods and results: The migratory response of both primary and subcultured 

rabbit Schwann cells was assessed using a modified Boyden chamber assay (McCarthy 
JB, et al., J Cell Biol 1983;97:772-777) (Baron-Van Evercooren A, et al., J Cell Biol 
1982;93:21 1-216). Schwann cells were detached from the tissue culture flasks with 
0.05% trypsin/EDTA (GIBCO) and resuspended at 3xlO^/ml in chemotaxis medium 

15 (DMEM w/ 0.5% BSA, Sigma) before being placed in the upper well of a 48-welI 
chemotaxis chamber (Neuroprobe). The lower wells of the chemotaxis chamber 
contained test reagents, which were reconstituted in chemotaxis medium. Upper and 
lower wells were separated by a 8-^m pore polyvinylpyrrolidone-free polycarbonate filter 
(Poretics), precoated with fibronectin (20 ng/ml). Chemotaxis chambers were incubated 

20 for 4 h at 37°C in a humidified atmosphere of air with 5% CO2. Thereafter, the filters 
were fixed and stained with Diff Qick (DADE). The filters were cut in half and mounted 
with the bottom side down (containing migrated cells) onto glass coverslips. Cells that 
had not migrated were removed from the upper surface with cotton swabs. After air 
drying, the coverslips were mounted onto glass slides and migration was quantified by 

25 counting migrated cells in 20 randomly selected high power (X400) fields. Each sample 
was assessed in quadruplicate, and results expressed as m±SEM chemotaxis index (CI, 
the ratio between the number of cells that migrated toward test substances and those 



wo 00/62798 



PCT/USOO/09765 



-55- 

which migrated toward medium control; migration toward medium reflects spontaneous 
migration). Zigmond-Hirsch checkerboard analysis (Zigmond SH, et al., J£xp Med 
1973;137:387-410) was performed by incubating the cells in the chamber with various 
doses of test substances either on the bottom of the filter (to establish a positive gradient), 
5 on top of the filter (to establish a reversed gradient), or on both sides of the filter in equal 
concentrations (to examine accelerated random movement). Statistical significance of 
data was determined by Mann- Whitney U-test and Kruskal- Wallis-test for nonparametric 
analysis of results. 

The chemotactic response of Schwann cells to graded concentrations of 

10 rhVEGFi65 protein was considered. VEGF stimulated migration in a dose-dependent 
manner, with maximal activity ranging firom 10 to 1000 ng/ml. At these concentrations, 
approximately 1.8 times more cells migrated through the polycarbonate filter in 
comparison to the media control. At the lowest concentration tested (1 ng/ml), 
rhVEGFi65 still induced a statistically significant increase in chemotaxis uith a CI of 

15 1.3±0.04. Nerve growth factor (NOP, Sigma) served as positive chemoattractant control 
(Anton ES,etal.,Proci\ra//^ca^ 5c/ 1/&4 1994;91:2795-2799). To check for the 
specificity of this VEGF-induced effect, we performed additional experiments using a 
neutralizing monoclonal anti-VEGF antibody (Sigma). Results demonstrate that the 
antibody (dilution 1:500) itself had no effect on Schwann cell migration. When the 

20 antibody was present together with rhVEGFies, we observed complete abrogation of the 
stimulatory effect of VEGF at 100 ng/ml as well as at 500 ng/ml were completely 
abrogated. 

The migratory response of Schwann cells to VEGF could be explained either by 
chemotaxis (directed movement of cells along a chemotactic gradient) or by 

25 chemokinesis (enhanced speed or fi-equency of random migration). To address this issue, 
we performed a series of checkerboard analyses (Example Table 2). Addition of VEGF 
exclusively to the upper compartment together with Schwann cells failed to enhance 
migration; in contrast, gradually increasing the concentration gradient of VEGF between 
the lower and upper compartment increased migration of Schwann cells toward the lower 

30 compartment. 

Example Table 2 Checkerboard Analysis of rh VEGFi6.s-induced Schwann 
Cell Migration. 



wo 00/62798 PCTAJSOO/09765 

-56- 





VEGF (ng/ml), Upper compartment 






0 


1 


100 


500 


VEGF(^g/ml) 


0 


15.93 ±1.1 


15.05 ±0.9 


14.03 ±0.9 




Lower 


1 


16.40 ± 1.2 


17.27 ±0.8 


17.02 ± 1.4 


16.43 ± 1.1 


Compartment 


100 


27.45 ± 0.8 


24.87 ±1.4 


16.81 ± 1.1 


17.93 ±0.7 




500 


33.90 ±1.2 


33.12 ±2.2 


27.56 ±1.9 


15.56 ± 1.6 



Conclusion: VEGF directly promotes Schwann cell chemotaxis. 

It should be understood that the preceding is merely a detailed description of 
certain preferred embodiments. It therefore should be apparent to those skilled in the art 
5 that various modifications and equivalents can be made without departing from the spirit 
and scope of the invention. The invention is intended to encompass all such 
modifications within the scope of the appended claims. 
EXAMPLE 5 : 

Methods and results: Diabetes was induced in female Sprague Dawley rats by i.p. 

10 streptozotocin (85 mg/kg) according to previously published methods. Blood glucose 
was monitored repeatedly and animals were deemed diabetic if blood glucose exceeded 
200 mg/dL. After a period of twelve weeks, the animals were treated with i.m. injections 
of either 250 micrograms of make plasmid DNA encoding for human VEGFies, or saline. 
The effects of treatment were assessed after four weeks by recording sciatic nerve motor 

15 and sensory conduction velocities (M. Kalichman et al., Brain Res. 1998, 810:130-137). 
The motor nerve conduction velocity (MCV) of the untreated, non-diabetic group was 
48.8 ± 3.0 m/s (mean ± SEM), and the sensory nerve conduction velocity (SCV) of this 
group was 62.0 ±4.9 m/s. The MCV of the saline-injected, diabetic group was 33,7 ±1 .3 
m/s and the SCV was 27.1 ± 2.2 m/s. Therefore, MCV as well as SCV were significantly 

20 slowed in the diabetic (saline-injected) rats, as compared to the non-diabetic control 

animals. However, when rats were injected with ph VEGFies, MCV was restored to 41.9 
±1.9 m/s, and SCV was improved to 59.5 ±7.0 m/s. Both MCV and SCV of the diabetic 
VEGF-treated group are significantly different from the corresponding parameters of the 
non-diabetic rats. Moreover, several non-diabetic rats were treated with ph VEGF) 65 as a 

25 control, and neither MCV nor SCV were affected by this treatment. 

Apfel et al. (Brain Res. 1994, 634:7-12) had reported that nerve growth factor 
(NGF) could ameliorate a sensory neuropathy caused by diabetes in streptozotocin- 



wo 00/62798 PCTA;S00/09765 

-57- 

treated rats. These investigators adrmnistered recombinant human NGF three times 
weekly at a dose of 3 mg/kg. After 1 1 weeks, the compoimd latency as a measure of the 
conduction velocity of the caudal nerve in the tail was measured. The imtreated diabetic 
rats had a mean compoimd latency of 1 .85 ± .09 m/s, while the NGF-treated diabetic rats 

5 had a mean compound latency of 1 .76 ±.04 m/s, and the control rats had a mean 

compound latency of 1 .40 ±0.03. The rats from the treated group exhibited better nerve 
function compared to the untreated group but their NCVs were still significantly lower 
than those of the control group. We found that i.m. ph VEGFies was able to completely 
restore both motor and sensory NCVs within four weeks. 

10 References: 

Kalichman MW, Dines KC, Bobik M, Mizisin AP, Nerve conduction velocity, 
laser doppler flow, and axonal caliber in galactose and streptozotocin diabetes, Brain Res, 
1998,810:130-137, 

Apfel SC, Arezzo JC, Brownlee M, Federoff H, Kessler JA, Nerve growth factor 
15 administration protects against experimental diabetic sensory neuropathy. 



20 



All references, patents and patent publications that are recited in this application 
are incorporated in their entirety herein by reference. 
What is claimed. 



wo 00/62798 PCTAJSOO/09765 

-58- 

CLAIMS 

1 . A method for treating a condition characterized by peripheral neuropathy 
in a subject comprising: 

administering at least one angiogenic growth factor to a subject in need of 
5 such treatment in an amount effective to alleviate a symptom of peripheral neuropathy in 
the subject, 

wherein the subject is otherwise free of symptoms calling for treatment 
with an angiogenic growth factor. 

2. A method for treating a condition characterized by peripheral neuropathy 
10 in a subject comprising: 

administering at least one angiogenic growth factor to a subject in need of 
such treatment in an amoxmt and for a period of time effective to alleviate a symptom of 
peripheral neuropathy in the subject, 

wherein the period of time is significantly greater than the period of time 
15 during which an angiogenic growth factor is administered to a subject for the purpose of 
revascularization of an ischemic tissue. 

3. A method for treating a condition characterized by peripheral neuropathy 
in a subject comprising: 

administering at least one angiogenic growth factor to a subject in need of 
20 such treatment in an amount effective to alleviate a symptom of peripheral neuropathy in 
the subject, 

wherein administering is by intramuscular injection into a tissue at an 
injection site that is proximate to a nerve suspected of a neuropathy and distal to an 
injection site that would be selected for revascularization. 

25 4. The method of claim 1 , 2, or 3, wherein the angiogenic growth factor is 

selected from the group consisting of a vascular endothelial cell growth factor (VEGF), 
an acidic fibroblast grov^ factor (aFGF), a basic fibroblast growth factor (bFGF), 
epidermal growth factor, transforming grovilh factor a and p, platelet-derived endothelial 
growth factor, platelet-derived growth factor, tumor necrosis factor a, a hepatocyte 

30 growrth factor (j catter factor), a colony stimulating factor (CSF), macrophage-CSF(M- 
CSF), granulocyte/macrophage CSF and nitric oxide synthase (NOS). 



wo 00/62798 PCT/USOO/09765 

-59- 

5. The method of claim 1, 2, or 3, wherein administering comprises 
administering a VEGF nucleic acid to the subject. 

6. The method of claim 1, 2, or 3, wherein administering comprises 
administering a VEGF nucleic acid and a bPGF nucleic acid to the subject. 

7. The method of claim 1, 2, or 3, wherein the angiogenic growth factor is a 
VEGF nucleic acid encoding a VEGF polypeptide selected from the group consisting of: 



(a) VEGF A 

(b) VEGFB 

(c) VEGF C 
10 (d)VEGFD 



(e)phVEGF,2,; 
(OphVEGFns; 

(g) phVEGFi65; and 

(h) phVEGF,89. 

8. The method of claim 1, 2, or 3, wherein administering is selected from the 
group consisting of intravenous administration, intra-arterial administration and 
intramuscular administration. 

9. The method of claim 1, 2, or 3, wherein administering is by intramuscular 
15 administration. 

10. The method of claim 1, 2, or 3, wherein the angiogenic growth factor is an 
angiogenic growth factor nucleic acid. 

1 1 . The method of claim 1, 2, or 3, wherein the angiogenic growth factor is an 
angiogenic growth factor polypeptide. 

20 12. The method of claim 1 1 , wherein the angiogenic growth factor is a VEGF 

polypeptide. 

13. The method of claim 1 1 , wherein the angiogenic growth factor is a VEGF 
polypeptide selected from the group consisting of: 



(a) VEGF A 
25 (b) VEGF B 

(c) VEGF C 

(d) VEGF D: 



(e)phVEGF,2i; 
(OphVEGFns; 

(g) phVEGFi65; and 

(h) phVEGF,89. 

14. The method of claim 1 , 2, or 3, wherein the condition is selected from the 
group consisting of: 
30 (a) an ischemic peripheral neuropathy; 

(b) a toxin-induced peripheral neuropathy; 

(c) a neuropathy associated with a systemic disease; and 



wo 00/62798 



PCTAJSOO/09765 



-60- 

(d) a genetically acquired peripheral neuropatiiy. 

1 5. The method of claim 1 , 2, or 3, wherein the condition is an ischemic 
peripheral neuropathy. 

1 6. The method of claim 1 5, wherein the ischemic peripheral neuropathy is a 
5 diabetic peripheral neuropathy. 

17. The method of claim 15, wherein the ischemic peripheral neuropathy is a 
peripheral vascular disease. 

18. The method of claim 14, wherein the peripheral neuropathy is a 
genetically acquired peripheral neuropathy. 

10 19. The method of claim 1 8, wherein the genetically acquired peripheral 

neuropathy is selected from the group of conditions consisting of peroneal muscular 
atrophy (Charcot-Marie-Tooth Disease, types I, II, and X), hereditary amyloid 
neuropathies, hereditary sensory neuropathy (type I and type II), porphyric neuropathy, 
hereditary liability to pressure palsy, Fabry's disease, adrenomyeloneuropathy, Riley-Day 

15 syndrome, Dejerine-Sottas neuropathy (hereditary motor-sensory neuropathy-III), 

Refsum's disease, ataxia-telangiectasia, hereditary tyrosinemia, anaphalipoproteinemia, 
abetalipoproteinemia, giant axonal neuropathy, metachromatic leukodystrophy, globoid 
cell leukodystrophy, Friedrich's ataxia, mononeuropathy multiplex, plexopathy, and pure 
motor neuropathy. 

20 20. The method of claim 14, wherein the peripheral neuropathy is a toxin- 

induced peripheral neuropathy. 

21 . The method of claim 20, wherein the toxin-induced peripheral neuropathy 
is a neurotoxicity mediated by an agent selected from the group consisting of a 
therapeutic agent, an antineoplastic agent, a food contaminant, an envirormiental 

25 pollutant. 

22. A method for treating a subject who has sustained a peripheral nerve 
injury, comprising: 

administering at least one angiogenic growth factor to a subject in need of 
such treatment in an amount effective to enhance peripheral nerve regeneration. 
30 23. The method of claim 22, wherein the subject is otherwise free of 

symptoms calling for treatment with an angiogenic growth factor. 



wo 00/62798 



PCT/US00/0976S 



I/I 



80 

70- 
60- 
50- 
40- 

30" 
20- 

lOH 

0 



Q Non- diabetic 
Q Diabetic 



n-s. 



* * 




/ 



Saline Saline VEGF 



SO- 
TO - 
60 
50- 
o 40- 

(A 

E 30H 
20- 
10- 



Q Non -diabetic 
Q Diabetic 



n.s. 



* * 



X 




A 



A 



Saline Saline VEGF 



FIG.IA 



FIG. IB